Sunteți pe pagina 1din 455

Landolt-Brnstein

Numerical Data and Functional Relationships in Science and Technology


New Series / Editor in Chief: W. Martienssen

Group VIII: Advanced Materials and Technologies


Volume 4

Radiological Protection

Editors:
A. Kaul, D. Becker
Authors:
D. Becker, G. Brix, A. Dalheimer, G. Dietze, H.R. Doerfel,
K.F. Eckerman, H. Graffunder, Y. Harima, K. Hayashi,
N. Ishigure, A. Kaul, H. Klewe-Nebenius, M. Lasch, H. Paretzke,
N. Petoussi-Henss, A. Phipps, H. Smith, J.W. Stather, G.N. Stradling,
D.M. Taylor, H.-G. Vogt, W. Weiss

ISSN 1619-4802 (Advanced Materials and Technologies)


ISBN 3-540-20207-2 Springer Berlin Heidelberg New York
Library of Congress Cataloging in Publication Data
Zahlenwerte und Funktionen aus Naturwissenschaften und Technik, Neue Serie
Editor in Chief: W. Martienssen
Vol. VIII/4: Editor: A. Kaul, D. Becker
At head of title: Landolt-Brnstein. Added t.p.: Numerical data and functional relationships in science and technology.
Tables chiefly in English.
Intended to supersede the Physikalisch-chemische Tabellen by H. Landolt and R. Brnstein of which the 6th ed. began publication in 1950 under title:
Zahlenwerte und Funktionen aus Physik, Chemie, Astronomie, Geophysik und Technik.
Vols. published after v. 1 of group I have imprint: Berlin, New York, Springer-Verlag
Includes bibliographies.
1. Physics--Tables. 2. Chemistry--Tables. 3. Engineering--Tables.
I. Brnstein, R. (Richard), 1852-1913. II. Landolt, H. (Hans), 1831-1910.
III. Physikalisch-chemische Tabellen. IV. Title: Numerical data and functional relationships in science and technology.
QC61.23 502'.12
62-53136
This work is subject to copyright. All rights are reserved, whether the whole or part of the material is concerned, specifically the rights of translation,
reprinting, reuse of illustrations, recitation, broadcasting, reproduction on microfilm or in other ways, and storage in data banks. Duplication of this
publication or parts thereof is permitted only under the provisions of the German Copyright Law of September 9, 1965, in its current version, and
permission for use must always be obtained from Springer-Verlag. Violations are liable for prosecution act under German Copyright Law.
Springer-Verlag Berlin Heidelberg New York
a member of BertelsmannSpringer Science+Business Media GmbH
Springer-Verlag Berlin Heidelberg 2005
Printed in Germany
The use of general descriptive names, registered names, trademarks, etc. in this publication does not imply, even in the absence of a specific statement,
that such names are exempt from the relevant protective laws and regulations and therefore free for general use.
Product Liability: The data and other information in this handbook have been carefully extracted and evaluated by experts from the original literature.
Furthermore, they have been checked for correctness by authors and the editorial staff before printing. Nevertheless, the publisher can give no
guarantee for the correctness of the data and information provided. In any individual case of application, the respective user must check the
correctness by consulting other relevant sources of information.
Cover layout: Erich Kirchner, Heidelberg
Typesetting: Authors and Redaktion Landolt-Brnstein, Darmstadt
Printing and Binding: AZ Druck, Kempten/Allgu
SPIN: 10723325

63/3020 - 5 4 3 2 1 0 Printed on acid-free paper

Preface
About 10 years before discovery of X-rays and natural radioactivity by W. C. Rntgen and H. Becquerel,
more precisely in 1883, Hans Landolt, Richard Brnstein and Julius Springer have started a series of
selected and easily retrievable physical data, which became a successful tool for natural scientists
working or practising a profession in fields of chemistry, physics or technology.
Now, i.e. about 120 years after start of this unique data collection and consequently about 100 years after
introduction of ionizing radiations and radionuclides in natural sciences, medicine and technology, the
Landolt-Brnstein New Series is submitting to the reader a full volume on protection of man against
ionizing radiations and radionuclides, i.e. Radiological Protection. A comparison with the 6th edition of
Landolt-Brnstein containing merely a six pages chapter on Strahlenschutz shows the rapid
development of the field within the last five decades.
Compared to many of the volumes in the Landolt-Brnstein Series published in the past, the present
publication in the group Advanced Materials and Technologies is not only a compilation of numerical
data and functional relationships for practical purposes. Rather a comprehensive accompanying text is
intended to impart to the scientific or professional user of Radiological Protection both data, the
concepts and scientific bases of the discipline devoted to prevention of health risks to man from exposure
to ionizing radiations and radionuclides.
Conceptually, radiological protection is based on the principles of justification of any use of ionizing
radiation, of optimization of the application of radiation, and limitation of the radiation risk to man and
his environment by acceptable doses, so that use of radiation and radionuclides in scientific research,
medicine, technique and daily life is always of net benefit to man.
Since findings of various scientific disciplines such as medicine, biology, biophysics, nuclear physics and
techniques are the basis for radiological protection, multidisciplinary knowledge of fundamentals of these
disciplines is necessary for an effective protection of man against health effects of ionizing radiations.
Consequently, the present volume contains contributions of experts internationally qualified in scientific
disciplines or subjects such as radiation physics, biology and medicine, external and internal dosimetry of
ionizing radiation and radionuclides, decontamination and decorporation of radionuclides, or physical
and biological measuring techniques. Although a previous volume in the Landolt-Brnstein Series has
already considered shielding against high energy radiation such as of accelerators or of cosmic origin the
specific item of assessment of radiation shielding was treated, too, however restricted to an extent being
necessary for completion of tasks of practical radiological protection, specifically in the field of lower
energies.
The present volume addresses to
those already working in radiological protection, under the aspect of making available to them
numerical data and functional relationships e.g. on assessment of radiation doses from external and
internal sources, or with the aim of further education and impartation of most recent knowledge in
radiological protection and scientific disciplines behind;
those participating in post-graduate education programmes in radiological protection with the aim to
get a qualified expert e.g. in medical radiation physics, or as an employee in a competent national
authority for health protection;
newcomers in the field of radiological protection to submit necessary knowledge on bases and
practices of this discipline;
advanced students of physics, techniques or medicine with special interest in a later professional
occupation as health physicists, engineers or technicians;
physicians practising in X-ray diagnostics, radiation oncology and nuclear medicine with special
interest in medical radiological protection.

In the hardcopy of the present volume a CD-ROM is included containing:


the full text in the multi-platform Adobe-Acrobat(pdf)-format with searchable fulltext index and
additional information and data, which would be beyond the scope of the printed version, within the
interactive programme SISy (for MS-Windows only). These refer e.g. to decay data of radionuclides
or normalized excretion functions for monitoring workers by quantitative assessment of intakes of
radionuclides and calculation of resulting doses.
For further numerical data such as dose coefficients for intake of radionuclides by workers or members of
the public that are available from publications e.g. of the International Commission on Radiological
Protection ICRP or of the International Commission on Radiation Units and Measurements ICRU either
as hardcopies or in the Internet are not contained on the CD-ROM. The reader is referred to the relevant
original sources.
The editors of the present volume want to thank the authors of the contributions for their careful work,
the Editor in Chief of the Landolt-Brnstein Series, Prof. Dr. W. Martienssen, for having put
"Radiological Protection" on the list of volumes to be prepared for the New Series, and the Publisher,
especially Drs. Ch. Meier and R. Poerschke from the editorial office for their permanent and very active
engagement in realizing the present opus.

The Editors

Braunschweig/Salzgitter, 2004

Contributors

Editors
D. Becker
Bundesamt fr Strahlenschutz
Fachbereich KT 2
Willy-Brandt-Strae 5
38226 Salzgitter-Lebenstedt
GERMANY

A. Kaul
Physikalisch-Technische Bundesanstalt
Bundesallee 100
38116 Braunschweig
GERMANY

Authors
D. Becker
Bundesamt fr Strahlenschutz
Fachbereich KT 2
Willy-Brandt-Strae 5
38226 Salzgitter-Lebenstedt
GERMANY
1 Intoduction

G. Brix
Bundesamt fr Strahlenschutz
Institut fr Strahlenhygiene
Ingolstdter Landstrae 1
Neuherberg
85764 Oberschleiheim
GERMANY
10 Measuring techniques

A. Dalheimer
Bundesamt fr Strahlenschutz
Fachbereich Strahlenschutz und Gesundheit
Kpenicker Allee 120-130
10318 Berlin
GERMANY
10 Measuring techniques

VII

VIII

Contributors

G. Dietze
Physikalisch-Technische Bundesanstalt
Abt. 6
Bundesallee 100
38116 Braunschweig
GERMANY
4 Radiological quantities and units
10 Measuring techniques

H.R. Doerfel
Forschungszentrum Karlsruhe
Hauptabteilung Sicherheit
Postfach 3640
76021 Karlsruhe
GERMANY
10 Measuring techniques

K.F. Eckerman
Health Sciences Research Division
Oak Ridge National Laboratory
1060 Commerce Park
Oak Ridge
Tennessee 37831-6480
USA
7 Internal dosimetry of radionuclides

H. Graffunder
Ingenieurbro Graffunder
Friedrichstrae 28
76297 Stutensee
GERMANY
Radiation Protection Information System (SISy)

Y. Harima
Tokyo Institute of Tecnology
Research Laboratory for Nuclear Reactors
7-3-4-307 Hikarigaoka
Nerima-ku
Tokyo
179-0072 JAPAN
5 Shielding against ionizing radiation

K. Hayashi
Hitachi, Ltd.
Nuclear Plant Engineering Department
Saiwai-cho, 3-1-1, Hitachi, Ibaraki,
317-8511 JAPAN
5 Shielding against ionizing radiation

Contributors

N. Ishigure
Research Center for Radiation Safety
National Institute of Radiological Science
4-9-1, Anagawa, Inage, Chiba
263-8555 JAPAN
7 Internal dosimetry of radionuclides

A. Kaul
Physikalisch-Technische Bundesanstalt
Bundesallee 100
38116 Braunschweig
GERMANY
1 Introduction
8 Decontamination
11 Exposures from natural and man-made radiation sources

H. Klewe-Nebenius
Forschungszentrum Karlsruhe
Institut fr Instrumentelle Analytik
Postfach 3640
76021 Karlsruhe
GERMANY
3 Physical fundamentals

M. Lasch
Kernkraftwerke Grundremmingen
Betriebsgesellschaft mbH
Postfach
89355 Grundremmingen
GERMANY
8 Decontamination

H.G. Paretzke
Institut fr Strahlenschutz
GSF-Forschungszentrum fr Umwelt
und Gesundheit, GmbH
Neuherberg, Postfach 11 29
85758 Oberschleiheim
GERMANY
6 External dosimetry

N. Petoussi-Henss
Institut fr Strahlenschutz
GSF-Forschungszentrum fr Umwelt
und Gesundheit, GmbH
Neuherberg, Postfach 11 29
85758 Oberschleiheim
GERMANY
6 External dosimetry

IX

Contributors

A. Phipps
National Radiological Protection Board
Chilton
Didcot
Oxfordshire OX11 ORQ
UNITED KINGDOM
7 Internal dosimetry of radionuclides

H. Smith
National Radiological Protection Board
Chilton
Didcot
Oxfordshire OX11 ORQ
UNITED KINGDOM
2 Biological effects of ionising radiation

J.W. Stather
National Radiological Protection Board
Chilton
Didcot
Oxfordshire OX11 ORQ
UNITED KINGDOM
2 Biological effects of ionising radiation
7 Internal dosimetry of radionuclides

G.N. Stradling
National Radiological Protection Board
Chilton
Didcot
Oxfordshire OX11 ORQ
UNITED KINGDOM
9 Decorporation of radionuclides

D.M. Taylor
5, Pan Poeth
Pen-y-bont, CF31 5BD
Wales
UNITED KINGDOM
9 Decorporation of radionuclides

H.-G. Vogt
Zentrum fr Strahlenschutz und Radiokologie
Universitt Hannover
Am Kleinen Felde 30
30167 Hannover
GERMANY
5 Shielding against ionizing radiation

Contributors

W. Weiss
Bundesamt fr Strahlenschutz
Fachbereich Strahlenhygiene
Institut fr Strahlenhygiene
Ingolstdter Landstr. 1
85764 Oberschleiheim
GERMANY
10 Measuring techniques

Landolt-Brnstein
Editorial Office
Gagernstr. 8, D-64283 Darmstadt, Germany
fax: +49 (6151) 171760
e-mail: lb@springer-sbm.com
Internet
http://www.landolt-boernstein.com

XI

Contents

XII

VIII/4 Radiological Protection


Contents
1

The development of the organizational and the conceptual basis of radiological


protection ........................................................................................................................................................................................................................ 1-1

2
2.1
2.2
2.2.1
2.2.2
2.2.3
2.2.4
2.3
2.3.1
2.3.2
2.3.3
2.3.4
2.4
2.4.1
2.4.2
2.4.3
2.4.4
2.4.5
2.4.6
2.4.7
2.5
2.5.1
2.5.2
2.6
2.6.1
2.6.2
2.6.3
2.6.4
2.7
2.8
2.9

Biological effects of ionising radiation .......................................................................................................................................... 2-1


Introduction .................................................................................................................................................................................................................... 2-1
Cellular effects ............................................................................................................................................................................................................ 2-2
Primary events following exposure to ionising radiation ................................................................................................ 2-2
Cellular damage and repair following the primary radiation events .................................................................... 2-3
Classification of radiation-induced damage .................................................................................................................................. 2-5
Implications of cellular damage for whole or partial body exposure .................................................................. 2-9
Deterministic effects.............................................................................................................................................................................................. 2-9
Tissue and organ development.................................................................................................................................................................... 2-9
Dose-response relationships for radiation damage ............................................................................................................. 2-10
Deterministic effects in humans following acute whole-body irradiation.................................................. 2-11
Deterministic effects following partial body irradiation............................................................................................... 2-12
Radiation-induced cancer ............................................................................................................................................................................. 2-15
Cancer development .......................................................................................................................................................................................... 2-15
Dose-response relationships ...................................................................................................................................................................... 2-16
Exposures to external radiation .............................................................................................................................................................. 2-18
Exposure to internally incorporated radionuclides ............................................................................................................. 2-20
Dose and dose rate effectiveness factors (DDREFs)........................................................................................................ 2-23
Risk coefficients for protection .............................................................................................................................................................. 2-23
Low dose studies ................................................................................................................................................................................................... 2-25
Hereditary disease ................................................................................................................................................................................................ 2-27
Categories of genetic damage .................................................................................................................................................................. 2-27
Risk coefficients for hereditary disease ......................................................................................................................................... 2-28
Irradiation in utero ............................................................................................................................................................................................... 2-29
Deterministic effects.......................................................................................................................................................................................... 2-30
Brain function........................................................................................................................................................................................................... 2-30
Risk coefficients for cancer........................................................................................................................................................................ 2-31
Hereditary disease ................................................................................................................................................................................................ 2-31
Summary of risk factors for cancer and hereditary disease ....................................................................................... 2-32
Conclusions ................................................................................................................................................................................................................ 2-32
References .................................................................................................................................................................................................................... 2-34

3
3.1
3.2
3.2.1
3.2.2
3.3
3.3.1
3.3.2
3.4
3.4.1
3.4.2
3.4.3
3.5

Physical fundamentals ..................................................................................................................................................................................... 3-1


Natural radioactivity .............................................................................................................................................................................................. 3-1
Elements, isotopes and radionuclides .................................................................................................................................................. 3-2
Atoms, electrons and the Periodic Table of Elements ........................................................................................................ 3-2
Atomic nuclei, nuclides and the Chart of Nuclides ............................................................................................................... 3-3
The structure of the atomic nucleus ....................................................................................................................................................... 3-4
Elementary particles .............................................................................................................................................................................................. 3-4
Nuclear transformations..................................................................................................................................................................................... 3-6
Radioactive decay ................................................................................................................................................................................................ 3-10
Basic properties ...................................................................................................................................................................................................... 3-10
Decay modes ............................................................................................................................................................................................................. 3-15
The natural radioactive decay families ........................................................................................................................................... 3-18
Radioactive radiation ........................................................................................................................................................................................ 3-24

Contents

XIII

3.5.1
3.5.2
3.6
3.6.1
3.6.2
3.6.3
3.6.4
3.6.5
3.6.6
3.7

Types of radiation ................................................................................................................................................................................................ 3-24


Physical properties of radiation.............................................................................................................................................................. 3-26
Nuclear fission and fission products ................................................................................................................................................. 3-30
Particle induced nuclear fission ............................................................................................................................................................. 3-30
Fission products ..................................................................................................................................................................................................... 3-30
Nuclear reactors ..................................................................................................................................................................................................... 3-32
Nuclear explosives .............................................................................................................................................................................................. 3-32
Radioactive inventory and nuclear waste..................................................................................................................................... 3-33
Release of radionuclides from the radioactive inventory of a nuclear reactor ....................................... 3-38
References .................................................................................................................................................................................................................... 3-39

4
4.1
4.2
4.2.1
4.2.2
4.3
4.3.1
4.3.2
4.3.3
4.3.4
4.4
4.4.1
4.4.2
4.5
4.5.1
4.5.2
4.5.3
4.6
4.6.1
4.6.2
4.7
4.8
4.9

Radiological quantities and units........................................................................................................................................................ 4-1


Introduction .................................................................................................................................................................................................................... 4-1
Radiation field quantities .................................................................................................................................................................................. 4-2
Scalar radiation field quantities .................................................................................................................................................................. 4-2
Vectorial radiation field quantities.......................................................................................................................................................... 4-4
Interaction coefficients and quantities................................................................................................................................................. 4-5
Cross section.................................................................................................................................................................................................................. 4-5
Mass attenuation coefficient and mass energy transfer coefficient ....................................................................... 4-5
Mass stopping power and linear energy transfer (LET).................................................................................................... 4-6
Mean energy expended in a gas per ion pair formed ........................................................................................................... 4-7
Quantities related to energy transfer ..................................................................................................................................................... 4-7
Stochastic quantities .............................................................................................................................................................................................. 4-7
Non-stochastic quantities.................................................................................................................................................................................. 4-8
Dose quantities in radiation protection ........................................................................................................................................... 4-10
Concept of radiation protection quantities .................................................................................................................................. 4-10
Protection quantities .......................................................................................................................................................................................... 4-11
Operational quantities ...................................................................................................................................................................................... 4-14
Radioactivity quantities .................................................................................................................................................................................. 4-18
Activity, specific activity, activity concentration, activity per area .................................................................. 4-19
Specific quantities for radon, thoron and their progeny................................................................................................ 4-19
Quantities for internal dosimetry .......................................................................................................................................................... 4-22
Limits, constraints, action levels ........................................................................................................................................................... 4-23
References .................................................................................................................................................................................................................... 4-27

5
5.1
5.2
5.3
5.3.1
5.3.2
5.4
5.4.1
5.4.2
5.4.3
5.5
5.5.1
5.5.2
5.5.3
5.5.4
5.6
5.7

Shielding against ionizing radiation ................................................................................................................................................ 5-1


Introduction .................................................................................................................................................................................................................... 5-1
Stopping power and range ............................................................................................................................................................................... 5-1
Penetration depths of charged particles ............................................................................................................................................. 5-2
Heavy charged particles ..................................................................................................................................................................................... 5-2
Electrons and positrons....................................................................................................................................................................................... 5-4
Photons ................................................................................................................................................................................................................................ 5-5
Basic shielding concept ...................................................................................................................................................................................... 5-5
Attenuation data of radioactive sources in shielding materials.................................................................................. 5-6
An example of the calculation of an ambient dose equivalent rate ....................................................................... 5-9
Neutrons ......................................................................................................................................................................................................................... 5-16
Basic shielding concepts ............................................................................................................................................................................... 5-16
Attenuation data of various neutron sources in shielding materials ................................................................. 5-16
Sample shield calculation ............................................................................................................................................................................. 5-18
Induced activity ...................................................................................................................................................................................................... 5-19
Computer codes and online nuclear data services............................................................................................................... 5-28
References .................................................................................................................................................................................................................... 5-32

XIV

Contents

6
6.1
6.1.1
6.1.2
6.2
6.2.1
6.2.2
6.2.3
6.2.4
6.3
6.3.1
6.3.2
6.4
6.5
6.5.1
6.5.2
6.6
6.7

External dosimetry .............................................................................................................................................................................................. 6-1


Protection and operational quantities ................................................................................................................................................... 6-1
Protection quantities .............................................................................................................................................................................................. 6-1
Operational Quantities ......................................................................................................................................................................................... 6-1
Dosimetric models ................................................................................................................................................................................................... 6-2
Models and phantoms of the human body ...................................................................................................................................... 6-2
Idealized geometries representing occupational exposures ........................................................................................... 6-4
Environmental source geometries............................................................................................................................................................ 6-4
Methods of calculating protection quantities in computational models............................................................ 6-5
Conversion coefficients for photons ..................................................................................................................................................... 6-6
Occupational .................................................................................................................................................................................................................. 6-6
Conversion coefficients for environmental gamma ray fields ................................................................................ 6-13
Conversion coefficients for neutrons ............................................................................................................................................... 6-20
Conversion coefficients for electrons .............................................................................................................................................. 6-21
Occupational exposure .................................................................................................................................................................................... 6-21
Environmental exposure ................................................................................................................................................................................ 6-23
Doses from external exposure of radionuclides in the environment ................................................................ 6-23
References .................................................................................................................................................................................................................... 6-42

7
7.1
7.2
7.2.1
7.2.2
7.2.3
7.2.4
7.2.5
7.2.6
7.2.7
7.2.8
7.3
7.3.1
7.3.2
7.3.3
7.3.4
7.3.5
7.3.6
7.4
7.4.1
7.4.2
7.4.3
7.5
7.5.1
7.6
7.6.1
7.6.2
7.6.3
7.6.4
7.6.5
7.7
7.7.1
7.7.2
7.8

Internal dosimetry of radionuclides ................................................................................................................................................ 7-1


Introduction .................................................................................................................................................................................................................... 7-1
Biokinetics of radionuclides in the body .......................................................................................................................................... 7-2
Inhalation .......................................................................................................................................................................................................................... 7-3
Ingestion......................................................................................................................................................................................................................... 7-10
Cuts and wounds ................................................................................................................................................................................................... 7-14
Absorption through intact skin ............................................................................................................................................................... 7-15
Systemic behaviour of radionuclides................................................................................................................................................ 7-15
Excretion ....................................................................................................................................................................................................................... 7-21
Embryo and foetus............................................................................................................................................................................................... 7-21
Transfer in maternal milk ............................................................................................................................................................................. 7-24
Dosimetric models ............................................................................................................................................................................................... 7-25
Introduction ................................................................................................................................................................................................................ 7-25
Absorbed fraction and specific absorbed fraction ............................................................................................................... 7-26
Computational models of the human anatomy ....................................................................................................................... 7-28
Dose rate per unit activity, S-factor ................................................................................................................................................... 7-31
Specific absorbed fractions for various radiations.............................................................................................................. 7-32
Calculation of doses to soft tissues and the skeleton........................................................................................................ 7-36
Dose coefficients ................................................................................................................................................................................................... 7-37
Method of calculation ...................................................................................................................................................................................... 7-38
Sources of dose coefficients ...................................................................................................................................................................... 7-41
Dose coefficients for selected radionuclides ............................................................................................................................ 7-43
Internal monitoring ............................................................................................................................................................................................. 7-46
Methods of individual monitoring....................................................................................................................................................... 7-47
Monitoring programme................................................................................................................................................................................... 7-51
Need for a monitoring programme ..................................................................................................................................................... 7-51
Routine monitoring ............................................................................................................................................................................................. 7-51
Special or task-related monitoring ...................................................................................................................................................... 7-52
Confirmatory monitoring .............................................................................................................................................................................. 7-52
Wound monitoring .............................................................................................................................................................................................. 7-52
Dose Assessment................................................................................................................................................................................................... 7-53
Estimation of intake and dose .................................................................................................................................................................. 7-53
Control of worker doses ................................................................................................................................................................................ 7-54
Monitoring data for radionuclides (H-3, Co-60, Sr-90, Ru-106, I-131, Cs-134, Cs-137, Ce-144
U-234, Pu-239, Am-241).............................................................................................................................................................................. 7-55
References .................................................................................................................................................................................................................... 7-68

7.9

Contents

XV

8
8.1
8.1.1
8.1.1.1
8.1.1.2
8.1.1.3
8.1.1.4
8.1.1.5
8.1.1.6
8.1.1.7
8.1.1.8
8.1.1.9
8.1.2
8.1.2.1
8.1.2.2
8.1.2.3
8.1.2.3
8.1.3
8.1.3.1
8.1.3.2
8.1.3.3
8.1.3.4
8.1.3.5
8.1.3.6
8.1.3.7
8.1.3.8
8.1.3.9
8.1.3.10
8.1.4
8.2
8.2.1
8.2.2
8.2.2.1
8.2.2.2
8.2.3
8.2.3.1
8.2.3.2
8.2.4
8.2.4.1
8.2.4.2
8.2.4.3
8.2.4.4
8.2.5
8.2.5.1
8.2.5.2
8.3

Decontamination .................................................................................................................................................................................................... 8-1


Decontamination of materials ...................................................................................................................................................................... 8-1
General approaches to decontamination ........................................................................................................................................... 8-2
Contamination .............................................................................................................................................................................................................. 8-2
Characteristics of oxide layer in BWRs and PWRs .............................................................................................................. 8-2
Other types of contamination ....................................................................................................................................................................... 8-3
Decontamination ....................................................................................................................................................................................................... 8-4
The use of decontamination in decommissioning ................................................................................................................... 8-4
Identification of decontaminable components ............................................................................................................................ 8-5
Effectiveness of decontamination, decontamination factor ........................................................................................... 8-6
Decontamination techniques (processes) ......................................................................................................................................... 8-7
Decontamination and secondary waste generation................................................................................................................. 8-8
Decontamination techniques for large volume closed systems ................................................................................. 8-8
Reactor decontamination in BWRs and PWRs .......................................................................................................................... 8-8
Fuel assemblies and decontamination.............................................................................................................................................. 8-12
Decontamination of sodium cooled systems............................................................................................................................. 8-12
Gas cooled reactors (WAGR) .................................................................................................................................................................. 8-13
Decontamination techniques for segmented parts ............................................................................................................... 8-13
Chemical decontamination .......................................................................................................................................................................... 8-13
Electrochemical decontamination ........................................................................................................................................................ 8-15
Jetting decontamination techniques ................................................................................................................................................... 8-18
Ultrasonic decontamination ....................................................................................................................................................................... 8-19
Decontamination by foams ......................................................................................................................................................................... 8-21
Decontamination by gels............................................................................................................................................................................... 8-21
Decontamination by pastes ......................................................................................................................................................................... 8-21
Mechanical decontamination techniques ...................................................................................................................................... 8-21
Decontamination by strippable coatings ....................................................................................................................................... 8-22
Melting ............................................................................................................................................................................................................................ 8-22
Decontamination techniques for building surfaces ............................................................................................................ 8-22
Decontamination of skin ............................................................................................................................................................................... 8-24
Introduction ................................................................................................................................................................................................................ 8-24
Transport of radioactive substances via the skin .................................................................................................................. 8-24
Anatomy of the skin........................................................................................................................................................................................... 8-24
Transport procedure ........................................................................................................................................................................................... 8-25
Skin dose at contamination ......................................................................................................................................................................... 8-25
Calculation of the equivalent dose to the skin......................................................................................................................... 8-25
Equivalent dose rate conversion coefficients ........................................................................................................................... 8-26
Decontamination measures ......................................................................................................................................................................... 8-30
Organisational and preliminary measures ................................................................................................................................... 8-30
First aid measures of skin decontamination............................................................................................................................... 8-30
Specific decontamination procedures .............................................................................................................................................. 8-31
Decontamination of specific body regions and organs .................................................................................................. 8-32
Procedure at residual contamination and fixing a reference value..................................................................... 8-32
Frequency of decontamination steps................................................................................................................................................. 8-32
Derivation of the reference value for residual contamination ................................................................................. 8-32
References .................................................................................................................................................................................................................... 8-34

9
9.1
9.2
9.2.1
9.2.2
9.2.3

Decorporation of radionuclides ............................................................................................................................................................ 9-1


Introduction .................................................................................................................................................................................................................... 9-2
General considerations ........................................................................................................................................................................................ 9-2
Factors affecting the efficacy of treatment ..................................................................................................................................... 9-2
Factors influencing treatment decisions ............................................................................................................................................ 9-3
Decision levels ............................................................................................................................................................................................................ 9-3

XVI

Contents

9.4.4
9.4.5
9.4.6
9.4.7
9.4.8
9.4.9
9.4.10
9.4.11
9.4.12
9.4.13
9.4.14
9.5
9.5.1
9.5.2
9.5.3
9.6
9.6.1
9.6.2
9.6.3
9.6.4
9.6.5
9.6.6
9.6.7
9.7
9.8

Perception of risk and its implications ................................................................................................................................................ 9-4


Approaches to treatment.................................................................................................................................................................................... 9-5
Methods of treatment ............................................................................................................................................................................................ 9-6
Non-specific procedures .................................................................................................................................................................................... 9-6
Procedures to enhance systemic radionuclide excretion................................................................................................... 9-7
General comments on the efficiacy of chelating agents for the actinides ....................................................... 9-8
What are the factors that govern the efficacy of chelating agents ? ..................................................................... 9-9
Can the efficacy of treatment be predicted from animal studies ? .......................................................................... 9-9
Are chelating agents always most effective when the radionuclides are present in circulating
blood ? ................................................................................................................................................................................................................................. 9-9
Is DTPA effective for all actinides ? ................................................................................................................................................ 9-10
Will the administration of chelating agents result in enhanced tissue deposition ? .......................... 9-10
Is the administration of sodium carbonate effective for uranium ? ................................................................... 9-10
Must chelating agents be administered promptly to be effective ? .................................................................... 9-10
Is intravenous injection the best mode of administration ?........................................................................................ 9-11
How can judgements on efficacy be made ?............................................................................................................................. 9-11
When should treatment start ? ................................................................................................................................................................. 9-11
When should treatment stop ? ................................................................................................................................................................. 9-11
For which materials are chelating agents likely to be effective ? ........................................................................ 9-11
For which materials are chelating agents unlikely to be effective ? ................................................................. 9-12
Is lung lavage more effective than chelation treatment for inhaled materials ?.................................... 9-12
Recent developments ........................................................................................................................................................................................ 9-13
Plutonium and americium ............................................................................................................................................................................ 9-13
Thorium .......................................................................................................................................................................................................................... 9-13
Uranium.......................................................................................................................................................................................................................... 9-13
Optimum treatment protocols................................................................................................................................................................... 9-14
Tritium ............................................................................................................................................................................................................................. 9-14
The alkaline earth elements, strontium, barium and radium..................................................................................... 9-14
Iodine ................................................................................................................................................................................................................................ 9-16
Caesium .......................................................................................................................................................................................................................... 9-16
Plutonium and americium ............................................................................................................................................................................ 9-18
Thorium .......................................................................................................................................................................................................................... 9-26
Uranium.......................................................................................................................................................................................................................... 9-27
Future research needs ....................................................................................................................................................................................... 9-29
References .................................................................................................................................................................................................................... 9-31

10
10.1
10.1.1
10.1.2
10.1.2.1
10.1.2.2
10.1.2.3
10.1.2.4
10.1.3
10.1.4
10.1.5
10.1.6
10.1.7
10.1.8
10.1.9
10.2
10.2.1

Measuring techniques .................................................................................................................................................................................. 10-1


Detectors for radiation protection ........................................................................................................................................................ 10-1
Overview and general characteristics of radiation detectors .................................................................................... 10-1
Gas-filled ionization detectors................................................................................................................................................................. 10-3
Ionization and gas amplification ........................................................................................................................................................... 10-3
Ionization chambers ........................................................................................................................................................................................... 10-4
Proportional counters........................................................................................................................................................................................ 10-8
Geiger-Mller counters ............................................................................................................................................................................... 10-10
Scintillation detectors .................................................................................................................................................................................... 10-11
Semiconductor detectors ............................................................................................................................................................................ 10-14
Thermoluminescence and radiophotoluminescence detectors ............................................................................. 10-18
Photographic films ........................................................................................................................................................................................... 10-20
Detectors for neutrons................................................................................................................................................................................... 10-21
Biological dosimetry ...................................................................................................................................................................................... 10-23
References for 10.1 .......................................................................................................................................................................................... 10-25
Radiological protection measurements: external exposure ..................................................................................... 10-27
Operational quantities ................................................................................................................................................................................... 10-27

9.2.4
9.2.5
9.3
9.3.1
9.3.2
9.4
9.4.1
9.4.2
9.4.3

Contents

XVII

10.2.2
10.2.3
10.2.3.1
10.2.3.2
10.2.4
10.2.4.1
10.2.4.2
10.2.4.3
10.2.5
10.2.5.1
10.2.5.2
10.2.6
10.2.6.1
10.2.6.2
10.2.6.3
10.2.6.4
10.2.7
10.3
10.3.1
10.3.1.1
10.3.1.2
10.3.1.3
10.3.1.4
10.3.2
10.3.2.1
10.3.2.2
10.3.2.3
10.3.2.4
10.3.2.5
10.3.2.6
10.3.2.7
10.3.2.8
10.3.2.9
10.3.2.10
10.3.2.11
10.3.3
10.3.3.1
10.3.3.2
10.3.3.3
10.3.3.4
10.3.3.5
10.3.3.6
10.3.3.7
10.3.3.8
10.3.3.9
10.3.3.10

Reference levels .................................................................................................................................................................................................. 10-29


Types of exposure ............................................................................................................................................................................................. 10-29
Occupational exposure ................................................................................................................................................................................. 10-30
Public exposure ................................................................................................................................................................................................... 10-30
Types of monitoring programs............................................................................................................................................................. 10-31
Individual monitoring for external exposure ......................................................................................................................... 10-31
Area monitoring for external exposure ....................................................................................................................................... 10-31
Calibration ................................................................................................................................................................................................................ 10-32
Requirements for individual monitoring of external exposure ........................................................................... 10-33
Operational requirements .......................................................................................................................................................................... 10-34
Accuracy requirement ................................................................................................................................................................................... 10-35
Personal dosimeters for individual monitoring in different radiation fields.......................................... 10-36
Photon dosimetry ............................................................................................................................................................................................... 10-36
Beta dosimetry ...................................................................................................................................................................................................... 10-37
Neutron dosimetry ............................................................................................................................................................................................ 10-37
Dosimetry in mixed field situations (photons and neutrons) ................................................................................ 10-38
References for 10.2 .......................................................................................................................................................................................... 10-39
Radiological protection measurements: internal exposure ...................................................................................... 10-41
Measurement of radon and its progeny ....................................................................................................................................... 10-41
Measurement of radon in air .................................................................................................................................................................. 10-44
Measurement of radon progeny in air........................................................................................................................................... 10-47
Measurement of radon in the ground and in water .......................................................................................................... 10-50
References for 10.3.1 ..................................................................................................................................................................................... 10-51
In vivo measurements ................................................................................................................................................................................... 10-52
Introduction ............................................................................................................................................................................................................. 10-52
Requirements ......................................................................................................................................................................................................... 10-53
Principles of spectrometry ................................................................................................................................................................... 10-56
Equipment ................................................................................................................................................................................................................. 10-58
Spectrum evaluation ....................................................................................................................................................................................... 10-66
Measuring geometries ................................................................................................................................................................................... 10-67
Calibration ................................................................................................................................................................................................................ 10-71
Uncertainties and detection limits..................................................................................................................................................... 10-77
Measurement procedure.............................................................................................................................................................................. 10-79
Quality assurance and control............................................................................................................................................................... 10-79
References for 10.3.2 ..................................................................................................................................................................................... 10-81
In vitro measurements: excretion analyses .............................................................................................................................. 10-83
Introduction ............................................................................................................................................................................................................. 10-83
Urine samples ........................................................................................................................................................................................................ 10-83
Faeces samples ..................................................................................................................................................................................................... 10-84
Exhalation ................................................................................................................................................................................................................. 10-84
Other biological samples............................................................................................................................................................................ 10-85
Radiochemical analyses .............................................................................................................................................................................. 10-85
Measuring techniques.................................................................................................................................................................................... 10-88
Quality assurance............................................................................................................................................................................................... 10-91
Examples for dose estimations from in vitro measurements ................................................................................. 10-93
References for 10.3.3 ..................................................................................................................................................................................... 10-97

11
11.1
11.2
11.2.1
11.2.2

Exposures from natural and man-made radiation sources ............................................................................. 11-1


Introduction ................................................................................................................................................................................................................ 11-1
Exposures by cosmic radiation and cosmogenic radionuclides............................................................................. 11-2
Origin and kinds of cosmic radiation ............................................................................................................................................... 11-2
Exposures by cosmic radiations............................................................................................................................................................. 11-2

XVIII
11.3
11.3.1
11.3.2
11.4
11.5
11.6

Contents
Terrestrial radiation ............................................................................................................................................................................................ 11-4
External exposures .............................................................................................................................................................................................. 11-5
Internal exposures ................................................................................................................................................................................................ 11-6
Enhanced exposures form industrial activities ....................................................................................................................... 11-9
Worldwide average exposure from natural and man-made sources .............................................................. 11-10
References ................................................................................................................................................................................................................. 11-12

1 The development of the organizational and conceptual basis of radiological protection

1-1

1 The development of the organizational and conceptual


basis of radiological protection1

Within a few weeks of Roentgen's discovery of X-rays in 1895, the potential of X-rays for diagnosing
fractures became apparent. However, the occurrence of acute adverse effects such as erythema and skin
burns within the next few years made persons applying X-rays in medicine and technique aware of the
need to avoid overexposure. Similar undesirable effects were reported after the discovery of natural
radioactivity by H. Becquerel in 1896, specifically of radium by M. Curie, and medical application.
The first organized coordinated effort for radiation protection came in 1921 when the British X-ray
and Radium Protection Committee issued detailed recommendations and instructions. The American
Roentgen Ray Society also proposed general recommendations in the early 1920s on the basis of avoiding
acute effects.
At the Second International Congress of Radiology held in Stockholm in 1928 [28B1], the unit
roentgen (R) was recognized as the unit for X-ray dose. It was at this congress that the International
X-ray and Radium Protection Commission was founded, the forerunner of the later (from 1950 onwards)
International Commission on Radioligical Protection (ICRP). The primary concern of the 1928
Commission was to elaborate recommendations designed to provide protection to members of the medical
profession in their work with X-rays and gamma-rays from radium. In 1934 the Commission
recommended 0.2 R per day as the tolerance dose [34I1].
Due to the great expansion in radiation protection work consequent upon nuclear energy
developments in the period from 1940 to 1950, the International Congress of Radiology in 1950 [51I1]
extended the scope of the Commission - now ICRP - and broadened its area of responsibility beyond the
protection of the medical profession only.
It was in 1950 that the ICRP spelt out the first time the various effects which were to be considered in
making its recommendations. These recommendations were to deal primarily with the basic principles of
radiation protection and to leave to the various international and regional agencies such as IAEA,
EURATOM and national regulatory bodies the responsibility of introducing detailed technical
regulations, codes of pratice or laws suited to the needs of their member countries or specific countries.
The present Commission of ICRP is assisted by 4 Committees working in the following specialized
fields [99I1]:
Committee 1 (Radiation Effects) considers the risk of induction of cancer and heritable disease
together with the underlying mechanisms of radiation action; also, the risks, severity, and mechanism
of induction of tissue/organ damage and developmental defects.
Committee 2 (Doses from Radiation Exposures) is concerned with the development of dose
coefficients for the assessment of internal and external radiation exposure, development of reference
biokinetic and dosimetric models, and reference data for workers and members of the public.

A concise and consolidated summary is given by A. Nagaratnam [95N1] in his handbook on the salient
features of the information given in ICRP Publications on the concept of radiological protection.
Landolt-Brnstein
New Series VIII/4

1-2

1 The development of the organizational and conceptual basis of radiological protection

Committee 3 (Protection in Medicine) is concerned with protection of persons and newborn children
when ionizing radiation is used for medical diagnosis, therapy, or for biomedical research; also,
assessment of the medical consequences of accidental exposures.
Committee 4 (Application of the Commission's Recommendations) is concerned with providing
advice on the application of the recommended system of protection in all its facets for occupational
and public exposure; it also acts as the major point of contact with other international organizations
and professional societies concerned with protection against ionizing radiation.
In its 1950 recommendations [51I1], ICRP replaced the 1934 concept of tolerance dose [34I1] by
that of the maximum permissible dose with the recognition that there could be risk even at these levels:
Whilst the values proposed for maximum permissible exposure are such as to involve a risk which is
small compared to other hazards of life, nevertheless, in view of the unsatisfactory nature of much of the
evidence on which our judgement must be based, coupled with the knowledge that certain radiation
effects are irreversible and cumulative, it is strongly recommended that every effort be made to reduce
exposure to all types of radiation to the lowest possible levels ... and that any unnecessary exposure be
avoided.
According to the 1958 recommendations [59I1] the basic permissible dose to gonads, bloodforming
organs, and lenses of the eyes for persons occupationally exposed at any age over 18 years was 5 rem
(50 mSv) per year or weekly 0.1 rem (1 mSv), used for purposes of planning and design. No
recommendation was made for exposure of individual members of the public but it was suggested that the
per capita dose should not exceed 5 rem (50 mSv) per generation excluding medical exposures and
exposures to natural background radiation. A linear non-threshold response was assumed for genetic
effects.
In 1962 it was recommended by the ICRP that the dose to individual members of the population at
large should be limited to 0.5 rem (5 mSv) per year [64I1]. ICRP made it explicit that doses from natural
background and from medical exposures were excluded from the maximum permissible doses. However,
ICRP recognizes especially the importance of the gonad doses resulting from medical exposure and the
attendant genetic hazard to the population, and recommended that the medical profession exercises
great care in the use of ionizing radiation in order that the gonad dose received by individuals before the
end of their reproductive periods be kept at the minimum value consistent with medical requirements.
In 1977 [77I1] ICRP published epoch-making recommendations giving a new philosophical and
conceptual framework of radiological protection. It is characterized by
1. Statement of the aim of radiation protection as being to prevent detrimental non-stochastic effects and
to limit the probability of stochastic effects to levels deemed to be acceptable.
2. Formulation of the basic tenets of the system of radiation protection as
a: Justification: No practice shall be adopted unless its introduction produces a positive net benefit.
b: Optimization: All (necessary) exposures shall be kept as low as reasonably achievable, economic
and social factors being taken into account (ALARA principle).
c: Dose limitation: The dose equivalents to individuals shall not exceed the limits recommended for
the appropriate circumstances by the Commission (limitation of the effective dose equivalent for
stochastic effects in workers to 50 mSv per year, for non-stochastic effects in specific organs to
500 mSv annually; limitation of the effective dose equivalent to control the risk from stochastic
effects of individual members of the public (critical groups) to 5 mSv in a year, and to 50 mSv
annually for non - stochastic effects).
Subsequent to the publication of the 1977 recommendations there have been clarifications and
amendments, the most important ones at the 1985 meeting of the Commission [85I1]:
Considering the effective dose equivalent limits for members of the public, made in its 1977
recommendations, the Commission's present view is that the principal (stochastic) limit is 1 mSv in a
year. However, it is permissible to use a subsidiary dose limit of 5 mSv in a year for some years, provided
that the average annual effective dose equivalent over a lifetime does not exceeed the principal limit of
1 mSv in a year.
Landolt-Brnstein
New Series VIII/4

1 The development of the organizational and conceptual basis of radiological protection

1-3

Apart from changes in terminology and definitions:

non - stochastic effects are now called deterministic effects,


quality factor is replaced by radiation weighting factor,
dose equivalent is replaced by equivalent dose,
effective dose equivalent is replaced by effective dose

the 1990 recommendations of the ICRP in its Publication 60 [91I1] have brought down significantly the
dose limits for occupational exposure from 50 mSv for the annual effective dose to 100 mSv in 5 years
corresponding to an average of 20 mSv annually. (Additionally the effective dose should not exceed 50
mSv in a single year).
The concept of justification, optimization and individual dose limits has been retained, however, a
distinction is made between the systems of protection for proposed and continuing practices, and
intervention:
While, as in the past, the system of protection in practices is following the general principles of
justification of a practice, optimization of protection (the magnitude of individual doses, the number of
people exposed, and the likelihood of incurring potential exposures should all be kept as low as
reasonably achievable, economic and social factors being taken into account), and limitation of individual
dose and risk, an additional system of protection in intervention has been introduced.
This is based on the following principles:
1. The proposed intervention should do more good than harm, i.e. the reduction in detriment resulting
from the reduction in dose should be sufficient to justify the harm and the costs, including social costs,
of the intervention.
2. The form, scale, and duration of the intervention should be optimized so that the net benefit of the
reduction of dose and consequently of the detriment should be maximized.
Regarding hereditary effects, ICRP Publ.26 [77I1] added the hereditary risk to the first and second
generation offspring to the stochastic risk to the exposed individual, the effects in later generations being
considered as part of the consequences for society. ICRP Publ.60 [91I1] now attributes the whole
hereditary detriment to the detriment suffered by the exposed individual, thus avoiding the need for a two
- stage assessment.
Under the motto Radiological Protection at the Start of the 21th Century ICRP in 2002 has started
an initiative which represents a genuine attempt to simplify the system of protection to one that is more
coherent and easily explicable [02C1; 02C2].
Since classical cost-benefit analysis based on an utilitarian ethical policy answering the question how
much does it cost to reduce a dose and how many lives are saved?, is unable to consider the individual,
the Commission already modified the principle of optimization by the introduction of the concept of a
constraint. Constraint is an individual-related criterion, applied to a single source in order to ensure that
the most exposed individuals are not subjected to excessive risk, and to limit the inequity introduced by
cost-benefit analysis.
Although in the future the process of taking all reasonable action to reduce exposures is still likely to
be called the Optimization of Protection, optimization is intended to be replaced by a different
requirement. Namely, residual doses, after the application of Constraints, should be kept as low as
reasonably achievable (ALARA). In this context the emphasis of constraints should provide a basic level
of health protection for individuals exposed to a particular controllable source. Since there is likely to be
some risk to health even at small doses introduction of a moral requirement is discussed for each
controllable source to take all reasonable steps to restrict both the individual doses to levels below the
action level and the number of exposed individuals. In this context it should be emphasized that these
Constraints are not intended to be applied to justified medical exposures.
Under the aspect common sense would be often more important than formal application of
differential equations in optimization stakeholder involvement is discussed to determine or negotiate for
the best level of protection in the circumstances. This means that whilst the dose constraints thus
represent a basic standard of individual health protection, stakeholder involvement determines how far
Landolt-Brnstein
New Series VIII/4

1-4

1 The development of the organizational and conceptual basis of radiological protection

below the action level is as low as reasonable practicable, and will avoid the previous formal costbenefit analyses. Consequently ALARA would represent the optimum level of protection from the source
under control or for an uncontrolled source.

References
28B1 Bureau of Standards, Circular No. 374: X-Ray and Radium Protection; Recommendations of the
2nd International Congress of Radiology, 1928; Br. J. Radiol.1, (1928), 359
34I1 International X-ray and Radium Protection Commission: Br. J. Radiol. 7, (1934), 1
51I1 International Commission on Radiological Protection: Radiology 56, (1951), 431; Br. J. Radiol.
24, (1951), 46
59I1 International Commission on Radiological Protection: Publ. 1, Pergamon Press, Oxford (1959)
64I1 Recommendations of the International Commission of Radiological Protection, as amended 1959
and revised 1962. ICRP Publ. 6. Pergamon Press, London (1964).
77I1 International Commission on Radiological Protection: Publ. 26, Annals of the ICRP 1 (3) (1977)
85I1 Statement from the Paris Meeting of the ICRP: Annals of the ICRP 15 (1985)
91I1 1990 Recommendations of the International Commission on Radiological Protection: Publ. 60,
Annals of the ICRP 21 (1991)
95N1 Nagaratnam, A.: Defence Research and Development Organisation, Ministry of Defense, New
Delhi - 110 011 (1995)
99I1 International Commission on Radiological Protection; Annual Report (1999): 24-06-2000
02C1 Clarke, R. H.: Int. Zeitschr. f. Kernenergie 47,1, (2002), 20
02C2 Clarke, R. H.: Strahlenschutzpraxis 8, 1, (2002), 45

Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

2-1

2 Biological effects of ionising radiation

This Chapter describes the effects of ionising radiation on the body. It covers both effects at the
subcellular and cellular level as well as on the whole body. Acute effects can result from high radiation
doses and in extreme cases can cause severe tissue damage and even death. For most exposures of people
to radiation it is low doses that are of most concern. These can give rise to radiation-induced cancer in
those exposed and hereditary disease in future generations. The chapter discusses the sources of
information on radiation damage and includes estimates of risk for these different effects.

2.1 Introduction
Within a few weeks of Wilhelm Conrad Roentgens discovery of X-rays on 8 November 1895, for which
he received the Nobel Prize for physics in 1901, the potential of the technique for diagnosing fractures
and other medical problems had become apparent, but acute adverse effects (such as hair loss, erythema
and dermatitis) were also found. Similar undesirable effects were reported shortly after the discovery of
radium (by Henri Becquerel in 1896) and its subsequent medical applications. In 1904, the first death of a
person exposed to X-rays was reported; X-ray burns had developed into cancer. This death was soon
followed by a steady stream of martyrs to science through roentgen rays to use the title of a book by a
radiologist who subsequently died of cancer. The widespread use of X-rays and radium in treating disease
in the early 1900s led to the recognition of a cancer risk in many organs and tissues following high
radiation doses which caused gross tissue damage. There was, however, a delay of about 40 years before
it became clear that there was a risk of radiation-induced cancer from irradiation at lower doses and that
there is no apparent threshold dose below which exposure to radiation can be considered safe. This delay
can be attributed to the fact that radiation-induced cancers do not differ in any known way from those
occurring naturally or caused by other agents. For many cancers there is also a long interval between
exposure and the appearance of the tumour. It is now believed that any radiation dose, whether from
external radiation or from incorporated radionuclides, is capable of inducing cancer and that the
probability of its occurrence, but not its severity, depends on the radiation dose. Animal studies have
shown that an increased incidence of certain types of inherited disorders can also occur in the descendants
of irradiated parents. For both cancer and inherited disorders the probability of their occurrence, but not
their severity, depends on the radiation dose. In radiological protection terminology they are termed
stochastic effects.
A second type of damage is seen after exposure of the whole or parts of the body to high doses of
radiation between a few gray and a few tens of gray. It is a reflection of impairment of the functional
capacity of tissues and is referred to as a deterministic effect. Severity of the damage is related to the
extent of radiation exposure and it is assumed that there is a threshold below which the clinically
detectable damage does not occur. If damage is extensive death may result. Following radiation exposure
in utero serious mental retardation has been observed in the children of the atomic bomb survivors in
Japan. Current evidence suggests this phenomenon is deterministic with a threshold related to the
minimum shift in intelligence quotient (IQ) that can be measured.
This Chapter reviews the sources of information available on the response of the body to radiation
damage, and considers the extent to which dose-response relationships can be determined and quantified.
Landolt-Brnstein
New Series VIII/4

2-2

2 Biological effects of ionising radiation

[Ref. p. 2-34

2.2 Cellular effects


2.2.1 Primary events following exposure to ionising radiation
Ionising radiations, hereafter abbreviated to radiation(s), can be classified into directly or indirectly
ionising. Charged particles such as alpha particles and beta particles emitted from radionuclides are
directly ionising if they have sufficient kinetic energy to disrupt atomic structure. Other types of radiation
such as X-rays (generated artificially) or gamma rays (from nuclear transitions) are indirectly ionising.
When passing through matter, they give up their energy to the atoms with which they collide and high
velocity charged electrons are ejected from these atoms leaving behind positive ions. These electrons
move randomly along a trajectory and may ionise other atoms in their path. If this occurs, more electrons
are ejected, while the incident electrons continue on their trajectory with decreased energy and velocity,
having transferred some of their energy to the newly formed electrons and eventually come to rest.
Neutrons also lose energy in various ways, an important means being through collisions with hydrogen
nuclei, which are single protons. The protons are set in motion and, being charged, they again deposit
energy through electrical interactions. The unique feature of ionising radiation, then, is the localised
release of energy in sufficient amounts to alter atomic and molecular structure.
The particle track is the ensemble of ionisations (and excitations) along the trajectory of the electron
or proton. One way of expressing the amount of atomic disruption is to quote the average energy loss
along the track. This is referred to as the unrestricted linear energy transfer (LET or L). LET quantities
are given in terms of average energy lost per unit path length, expressed in terms of kiloelectronvolts per
micron (keV m1). This physical quantity has been used extensively in experimental radiobiology as a
simplistic approach in order to relate the quality of radiation to cellular damage [91I2].
The rate of energy loss in biological material can vary greatly along the particle track depending upon
kinetic energy and charge. In general terms, photons and electrons have LET values in the range of about
0.2 to 10 keV m1; for example, 1 MeV, 100 keV, 10 keV and 1 keV electrons have LET values of 0.2,
0.5, 2, and 10 keV m1 respectively. Protons, alpha particles and neutrons have LET values between
about 10 and 100 keV m1; and heavy charged particles (e.g. nuclei of elements such as C, Ne and Si)
can have still higher values to about 2000 keV m1.
LET does not address the magnitude of the individual energy-loss events that occur along the track;
nor does it address the amount of energy lost to matter in the volume of interest. This can be expressed as
mean lineal energy which, in concept is more meaningful than LET [93I4].
The random nature of the particle track can be simulated by computer analysis using Monte Carlo
techniques. A two-dimensional clustering of ionisations is shown in Fig. 2.1. This is only an
approximation of the more complex three-dimensional events that involve random clustering of
ionisations on a sub-atomic scale. Nevertheless, the figure illustrates the concept that low energy
electrons are sparsely ionising because the ionisations are well separated spatially. Alpha particles, in
contrast, are densely ionising because the ionisations are closely packed together along the track. It has
been calculated that a single particle track of low-LET radiation (e.g. 1 MeV gamma-rays) passing
through an 8 m diameter spherical nucleus delivers an absorbed dose of about 1 mGy [94G2]. The
gamma-rays are about one hundred times less damaging than high-LET radiation, for example 1 MeV
neutrons which deliver an absorbed dose of a few hundred mGy in the same shape of nucleus.
Each ionisation can result in energy being deposited within the atoms of a target molecule in sufficient
amounts to disrupt chemical bonds. Alternatively, it may indirectly break the chemical bonds in a nearby
molecule. It is the predominant reaction in water molecules in cells after exposure to X-rays. Free
hydroxyl and other related radicals are produced and during their short existence of about a microsecond,
these highly reactive radicals are capable of diffusing a few micrometres to reach and damage a target
molecule such as deoxyribonucleic acid (DNA).

Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

2-3

Fig. 2.1. Simulated low energy


electron track (upper: initial
energy 500 eV) and simulated
short portion of an alpha particle
track (lower: 4 MeV). Large
circles are ionisations, small
circles are excitations. A Section
of DNA is shown to give a
perspective
on
dimensions;
[94G2].

2 nm

The temporal sequence of ionisations (and excitations) leading to biological effects is illustrated in
Fig. 2.2. Physico-chemical events are completed rapidly, the repair of damage may be completed within
tens of minutes while effects in cells can arise within hours or days. The biological manifestations in
multi-cellular organisms, including man, can be delayed for many years or, as in the case of hereditary
disease, only be manifest in future generations.
Tissue and whole body effects
Cellular effects

Enzymatic actions
( repair/fixation of damage )
Formation of radicals
and radical interactions
Ionisations and excitations

1 min

1 year

Fig. 2.2. Timescale of events


leading to radiation effects
following exposure to ionising
radiations.

Seconds

2.2.2 Cellular damage and repair following the primary radiation events
It is widely accepted that the most important cellular constituent to be damaged by radiation is nuclear
DNA. The molecular structure consists of a double helix (Fig. 2.1), formed from two complementary
strands of nucleotides. These are purine and pyrimidine bases linked to sugar molecules with phosphate
Landolt-Brnstein
New Series VIII/4

2-4

2 Biological effects of ionising radiation

[Ref. p. 2-34

molecules joined by ester linkages. The two strands are held together by hydrogen bonds between
guanine-cytosine and adenine-thymine base pairs. The cells genetic information is carried in a linear
sequence of nucleotides that make up the estimated 100,000 genes in the human genome. Each gene
controls a discrete characteristic.
Just as cells inherit genes, they also inherit a set of instructions that tell the genes when to become
active. These gene regulatory proteins recognise short stretches of nucleotide sequences on the double
helix and determine which of the genes in the cell will be transcribed. About two-thirds of genes provide
instructions for cell division and for the synthesis of tens of thousands of proteins that provide the
structural components of cells, as well as numerous enzymes promoting and controlling cellular activity.
Ribonucleic acid (RNA) is the molecule that helps to transport, translate and implement the coded
instructions from the genes in the nucleus to the body of the cell. All cell types contain the same genes,
but encoding sets of genes is cell-specific. This uniqueness ensures that cells in each tissue produce their
own proteins.
Maintaining stability in the gene is essential for cell survival. This stability requires not only
extremely accurate mechanisms for DNA synthesis and replication, but also mechanisms for repairing
DNA damage before replication. Observations with proliferating cells in the laboratory indicate that DNA
is subjected to only an estimated few tens of base-pair or nucleotide permanent changes per year during
normal metabolism, despite the fact that metabolic processes alter thousands of bases and nucleotides
every day.
DNA single strand breaks, without base involvement, are effectively ligated enzymatically. Base
excision repair pathways require different groups of enzymes that identify and excise the damaged base
site, make a complementary copy of the information bases on the opposite undamaged strand, and seal the
correct sequence of copied bases in the gap. If nucleotide damage occurs, nucleotide excision repair
pathways are able to repair the more extensive damage on one strand. Once the lesion is identified along
the strand, the damaged nucleotides are removed and repair proceeds thereafter as for base damage.
DNA double strand damage with or without base damage, occurs much less frequently than damage to
single strands during normal cellular activity. Recombination repair pathways are available, but they are
not totally effective, since there is no undamaged strand to act as a template for base or nucleotide
replacement. Damage to bases can result in their alteration or loss. When the repair processes fail, the
resulting misrepair is referred to as a mutation.
DNA damage due to radiation causes similar lesions to those occurring after normal metabolism, but
double strand breaks, multiple gene losses and the translocation of gene sequences occur more frequently
as a dose-related effect. The probability of misrepair is greater under these circumstances. Estimated
yields of damage caused by low-LET radiation are shown in Table 2.1 [88W1].
Table 2.1. Examples of damage in a mammalian cell nucleus from 1 Gy of low-LET radiation.
Initial physical damage
Ionisations in cell nucleus
~ 100,000
Ionisations directly in DNA
~ 2,000
Excitations directly in DNA
~ 2,000
Selected biochemical damage
DNA single-strand breaks
~ 1,000
Base (8-hydroxyadenine) damage
~ 700
Base (thymine) damage
~ 250
DNA double-strand breaks
~ 40
DNA-protein cross links
~ 150
Selected biochemical damage
Lethal events
~ 0.2-0.8
Chromosome aberrations
~ 0.4
Hprt(1) gene mutations
0.6 105
Translocation frequency (2 loci)
1.2 104
(modified from 88W1)
(1) hypoxanthine-phosphoribosyl transferase
Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

2-5

Recent investigations have revealed that DNA repair pathways may work in conjunction with other
intracellular activities in order to minimise cell damage. These include delay in cell-cycling (as a means
of maximising the chances of repair); and programmed cell death (apoptosis), whereby severely damaged
cells are eliminated to stimulate cell proliferation.

2.2.3 Classification of radiation-induced damage


Laboratory techniques have been available for many years to observe radiation effects in proliferating
cells. These techniques include measuring changes in cell survival, in the frequency of chromosomal
aberrations (deletions and translocations), in gene structure (mutations), and in oncogenic transformation
(neoplasia).
2.2.3.1 Cell survival
Cellular damage can be classified into three arbitrary categories: lethal damage which results in cell
death; sublethal damage, which may be repaired; and potentially lethal damage, defined as damage that
can be repaired by altering the growth conditions as for cells in culture.
Cell lines of fibroblasts from rodent and human tissues have been used extensively to establish doseresponse relationships [93U6]. Expressed graphically as the logarithm of cell survival plotted against
absorbed dose on a linear scale, the dose-response is linear for low-LET radiation at low doses, followed
by a curvature at higher doses.
Expressed mathematically, the relationship can be represented by a linear-quadratic equation:
S = e ( D+ D

(1)

where S is the surviving fraction after exposure to dose D and and are coefficients representing the
linear and quadratic components for cell killing. The initial slope of the relationship is determined by ,
while the quadratic component, reflects the curvature in the dose-survival relationship (Fig. 2.3). The
dose at which the linear and quadratic components are equal is the ratio of and . The response to highLET radiation is also shown in Fig. 2.3 where survival is best expressed as a linear function of dose
passing through the origin.
1
aD
b D2

Cell survival

10 -1

10 -2
High
LET
10 -3
0
Landolt-Brnstein
New Series VIII/4

Low
LET

a/b
4

8
Dose D [Gy]

12

16

Fig. 2.3. Typical survival curves for cultured cells


exposed at high dose-rate (>0.1 Gy min1). The curves
illustrate the linear-quadratic relationship for low-LET
radiations and linear relationship for high-LET
radiations.

2-6

2 Biological effects of ionising radiation

[Ref. p. 2-34

A plausible explanation of the linear component following exposure to low-LET radiation at low
doses is that the majority of DNA interactions are single particle track events [94G2]. Under these
circumstances, DNA damage can be effectively repaired. As the dose increases, multi-track events
reflecting the quadratic component, and which are associated with clustered DNA damage, increasingly
predominate with a consequent increase in the probability of misrepair and lethal events. At 1 Gy, for
example, lethal events have a frequency of about 0.2 to 0.8 per cell (Table 2.1).
Protracted exposure to low-LET radiation results in less damage, per unit of dose, compared with
acute exposure [93U6, 00U8]. This is referred to as the dose rate effect and is due to the ability of cells to
repair more sublethal damage as the dose rate is reduced. Below about 1 Gy min1, the slope on the
exponential portion of the survival curve typically becomes progressively shallower as more and more
sublethal damage is repaired. Below about 0.01 Gy min1, undamaged cells are able to proliferate at a
sufficient rate to offset the reduction in cell numbers while repair is progressing. This response is
illustrated in Fig. 2.4. A dose rate effect is not observed after exposure to high-LET radiation, suggesting
little repair of damage.
1

Surviving fraction

0.004 Gy min -1
10 -1
Proliferation

0.01 Gy min -1

10 -2

Repair

1 Gy min -1

10 -3
0

8
Dose D [Gy]

12

16

Fig. 2.4. Dose-rate effect showing the influence of


repair and repopulation on the dose-survival relationship
for cells.

The relative biological effectiveness (RBE) of different types of radiation is defined as the ratio of a
dose of a reference low-LET radiation to a dose of the test radiation that gives an identical biological
endpoint [90N3]. RBE values are influenced by variations in LET, dose and dose rate. RBE values
increase to a maximum at about 100 keV m1, decreasing thereafter because of an overkill effect. The
absolute value of the RBE is not unique but depends on the level of biological damage and, therefore, on
the absorbed dose [86B1]. For irradiation by alpha particles, for example, the RBE is generally taken to
be 20 for stochastic effects (cancer and hereditary disease) but to have a lower value of around 5 for
deterministic effects.
2.2.3.2 Damage to viable cells
Chromosome aberrations and gene mutations
The technique of culturing human lymphocytes in vitro has been available for many years. It provides a
means of measuring the frequency of unstable and stable chromosome aberrations at various stages in the
cell-cycle. In terms of unstable aberrations, their frequency increases from a background level of about
103 to a rate of about 4 102 Gy1 after exposure to low-LET radiations. Dose-response relationships
for different types of radiation are illustrated in Fig. 2.5 [89E1]. Neutrons are more damaging than X-rays
Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

2-7

or gamma-rays and low energy neutrons are more damaging than high energy neutrons. For low-LET
radiations a linear-quadratic relationship is consistent with the data.
That is:
E = D +D2

(2)

where E is the frequency of chromosome aberrations (i.e. a stochastic effect), D is the dose, and and
are the linear and quadratic coefficients for the induction of the aberrations.
1.0

Dicentrics per cell

Fission neutrons

0.5

250 kVp X - rays

0
0

Fig. 2.5. Dicentric yield in chromosomes per cultured


human lymphocyte as a function of dose for selected
radiations; [89E1].

Dose D [Gy]

A number of specific-locus mutation test systems using mouse, hamster and human fibroblasts have
been developed to measure mutagenesis. One cell line, the human B-lymphoblastoid TK6, illustrates the
use of the test [89K4]. Cultured cells were exposed to radiation and the mutation frequency at two loci
(hgprt and tk) was measured under different exposure conditions. For acute radiation exposure, 100 kVp
X-rays (0-2 Gy) and (Pu, Be) neutrons (0-0.2 Gy) both showed a linear dose-response relationship in
terms of induced mutants. The induced mutant frequency per 0.01 Gy per surviving cell was 0.55 l07
(CI 0.09) and 1.92 l07 (CI 0.03) respectively.
Protracted exposure to X-rays (0.01-0.1 Gy per day) for 5 to 20 days showed a slight increase in the
mutation frequency (0.84 l07 (CI 0.17)); while continuous exposure to neutrons (0-0.4 Gy) resulted in a
substantial increase (6.00 l07 (CI 0.7)). These data demonstrate an inverse dose-rate effect for
neutron-induced mutation in human cells. Syrian hamster embryo cells showed a similar effect, but other
cell lines did not. It is concluded that there are a number of difficulties in interpreting the results of
somatic cell mutations.
Estimated yields of chromosome aberrations and mutation frequency are shown in Table 2.1.
Cell transformation
An established technique for studying carcinogenic potential is that of culturing cells that can grow
indefinitely, provided that they are frequently transferred to fresh media. Under specific conditions, cells
that have acquired this ability are said to be immortalised. A characteristic of these immortalised cells is
that they stop dividing when they come into contact with similar cells in the culture medium (contact
inhibition). They are not classified as malignant cells because they do not cause tumours when injected
into immunologically-suppressed animals. Occasionally, an immortalised cell undergoes a spontaneous
change, whereby it loses its contact inhibition and continues to proliferate by spreading over adjacent
immortalised cells to form a recognised foci of cells. Such cells are said to have undergone
Landolt-Brnstein
New Series VIII/4

2-8

2 Biological effects of ionising radiation

[Ref. p. 2-34

transformation and when they are injected into animals, they develop into tumours. Spontaneous
transformation is a rare event, occurring at a rate of about one in ten thousand to one in a hundred
thousand per surviving cell. The mechanism is not fully understood, but it is thought to involve the
mutation of two or more genes. Two classes of mutated genes in particular have been identified and
characterised. These are gain-of function mutations of proto-oncogenes, whereby the mutated genes
(oncogenes) stimulate cell proliferation in an uncontrolled manner; and loss-of function tumour
suppressor genes, whereby cells are no longer prevented from proliferating in defiance of normal
controls.
The cell types used in transformation studies are mainly derived from fibroblasts. It is generally
accepted that the sensitivity of the test is low, the detection limit being about 0.25 Gy. Ideally, human
epithelial cells would be a better choice to represent human cancers. Future studies are in hand which aim
to use this type of cell.
To illustrate the technique, C3H/1OT fibroblasts derived from the prostate of the C3H mouse
embryo, were irradiated with low-LET radiation [98M2]. Cell survival and transformation frequencies
were simultaneously measured. The survival curve was consistent with a linear-quadratic dose-response
relationship (Equation 1), while the transformation frequency per surviving cell following exposure to
X-rays was consistent with a linear relationship (Fig. 2.6a). However, if the number of transformants per
cell at risk was plotted, the relationship to intermediate doses was consistent with a linear-quadratic
equation, the transformation frequency reaching a maximum at about 2 Gy (Fig. 2.6b). This doseresponse relationship is consistent with other results reported in the literature, although the maximum
transformation frequency was usually in the 3 to 4 Gy range.
6

2.0

Transformation frequency [10 -4 ]

Transformation frequency [10 -4 ]

5
4
3
2
1

1.6

1.2

0.8

0.4
Cells at risk

Cells viable for survival


0
0

2
3
4
Absorbed dose D [Gy]

2
3
4
Absorbed dose D [Gy]

Fig. 2.6. Transformation frequencies per surviving C3H 10 T cell (a) and per cell at risk (b) as a function of
absorbed dose after exposure to 250 kVp X-rays at 2 Gy min1; [98M2].

Exposure to neutrons resulted in a higher transformation frequency than for low-LET radiation, with
no evidence of a dose rate effect. One exception was a study of 5.9 MeV or fission neutrons where an
inverse dose-rate effect was reported [93U6]. It is concluded that there are difficulties in interpreting data
on cell transformation studies.
Generalised dose-response relationships
The conventional approach to representing the absolute biological effectiveness of a given radiation at
low doses is based on the assumption derived from target theory in which the induction I of an effect as a
function of dose D can be represented by
Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

I (D )= 1 D + 1 D 2 e ( 2 D+ 2 D

2-9

(3)

in which 1 and 1 are single and multihit components for a radiation effect and 2 and 2 represent single
and multihit components for cell killing. At low doses the incidence from effect is determined by 1 with
the response increasing linearly with dose. It is generally assumed that in this region 1 will be
independent of dose rate. With increasing dose the amount of damage due to multihit effects increases,
resulting in a quadratic component in the dose-response curve. At doses above a few gray 1 and 2
become significant resulting in a reduction in tumour yield due to the effect of cell killing.
For high-LET radiation ( particles, neutrons) the dose-response curve is generally found to be linear
up to the point at which cell killing starts to exert an effect and reduces the tumour yield.

2.2.4 Implications of cellular damage for whole or partial body exposure


The outcome of cell damage in terms of human radiation detriment can be profoundly different according
to the exposure conditions. Cellular studies can provide a sound basis upon which to judge these
outcomes. After acute exposure to absorbed doses above a few gray, the cells at greatest risk are selfreplicating stem cells that supply functional cells. They are programmed to divide so that one daughter
cell remains as a stem cell (in order to ensure that stem cell numbers in the tissue remain constant), while
the other daughter cell proceeds to specialise (differentiate) by clonal expansion. If sufficient numbers of
stem cells in a tissue are killed or are prevented from dividing at the appropriate rate, the tissue loses its
ability to function effectively. The consequential effects are referred to as deterministic. Studies have
established that cell survival is dose and dose-rate dependent for low-LET radiations, and that there is a
tissue-specific dose threshold. At high risk are rapidly dividing bone marrow stem cells, and stem cells in
the epithelium of the gastrointestinal tract, lungs, thyroid, gonads, skin and lens of the eye.
The effects due to the proliferation of mutated cells at low doses are termed stochastic. There is
sufficient radiobiological evidence for low-LET radiation to support the general assumption of an
increasing risk of an effect with increasing dose at low to intermediate doses, with no threshold. Cellular
techniques are providing insight into the way in which radiation can initiate the complex multistage
process of carcinogenesis. However, there is still much to be learned about the molecular changes that
lead to cells with the potential towards malignancy; and most importantly, any advances in knowledge at
the cellular level have to be seen in the context of the living organism.

2.3 Deterministic effects


2.3.1 Tissue and organ development
In the space of a few weeks, a single fertilised human egg gives rise to a complex multicellular organism
consisting of embryonic cells arranged in a precise pattern, each in its proper place. In the subsequent
period of fetal growth, the cells continue to proliferate in the developing tissues and organs. Growth of
tissues and organs continues in childhood with increase in cell mass in many tissues, but growth
essentially ceases in the adult when cell masses reach a predetermined size.
The majority of cells in tissues of the adult are differentiated, that is, they have developed specific
morphology and function which is usually irreversible, but these cells are predestined to die. In many
tissues of the body, the rate of death of differentiated cells is rapid and, in a healthy state, must be
balanced by proliferation from stem cells. These cells, by definition, are cells that have retained
Landolt-Brnstein
New Series VIII/4

2-10

2 Biological effects of ionising radiation

[Ref. p. 2-34

embryonic characteristics. They are able to divide during the lifetime of the organism, yielding progeny
that are destined to differentiate by a process of clonal expansion. Stem cells also retain the ability of selfrenewal. These characteristics are illustrated in Fig. 2.7. The number of stem cells compared to
differentiated cells varies according to the tissue, but they usually represent, at most, a few percent of the
total cell numbers. Furthermore, only a small fraction of the stem cells are active at any one time under
normal circumstances. It is not known how the balance between cell proliferation and cell death is
achieved, but it is thought that all cells are genetically programmed to die, by apoptosis. When
differentiated cells die, a feed-back mechanism is activated to stimulate the stem cells to divide and
replenish the population.

Stem cell

Self - renewal

Clonal
expansion
Maturation
without division
Functional cells with finite lifetime

Fig. 2.7. Derivation of differentiated cells from a selfrenewing stem cell.

If enough stem cells in a tissue are killed or prevented from undergoing cell division, there will be loss
of tissue function; termed deterministic by the International Commission on Radiological Protection
(ICRP). The dose-response relationship is characterised by a frequency and severity that increases with
dose above a threshold. Most tissues and organs of the body are able to compensate for small reductions
in the number of differentiated cells. But if the decrease is large enough, there will be changes seen as
loss of tissue or organ function and a consequential response to repair the damage.

2.3.2 Dose-response relationships for radiation damage


The probability of detecting loss of tissue or organ function following exposure to radiation increases
steeply above a threshold dose to a maximum. Expressed as a generalised dose-response relationship, the
plot of the frequency of the effect versus dose expressed on linear axes is sigmoid (Fig. 2.8, upper panel).
Above the threshold dose, the severity of the effect also increases with dose reflecting more cell loss and
hence damage to tissue function (Fig. 2.8, lower panel). Protracting the dose results in a lower frequency
and less severe symptoms at a given dose compared with acute exposure, demonstrating the importance
of stem cell repopulation.
There is individual variation in radiosensitivity in any exposed population. This variation reflects
differences in the ability of individuals to cope with radiation-induced cellular damage. Any response is
influenced by the age and state of health of the exposed individual.

Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

2-11

Frequency [%]

100

50

Seveity

Variation
in
population

Threshold

Fig. 2.8. Dose-response relationship for deterministic


effects. Variation in frequency and severity; [based on
91I2].
Dose

2.3.3 Deterministic effects in humans following acute whole-body irradiation


Evidence of the deterministic effects of radiation comes from several sources. These include retrospective
studies on radiotherapy patients, radiologists in the early part of the 20th century, Japanese populations
exposed to radiation from atom bombs, and individuals accidentally exposed to high doses following
nuclear reactor accidents and radiographic sources. Understanding the effects of acute high doses is
important as an aid to prognosis in the treatment of accidental over-exposure, and to ensure that
deterministic effects are avoided in normal practices and minimised in accidents. Evidence on
deterministic effects also comes from studies with animals.
After exposure to doses of a few Gy, the depression in the numbers of circulating white blood cells
(granulocytes) and blood platelets may be so severe as to result in death from septicaemia (infection) and
haemorrhage. This is referred to as the haematopoietic syndrome. Recovery depends upon the radiation
dose and the ability of the remaining stem cells in the marrow to recover. Loss and recovery of
granulocytes and blood platelets follows a similar dose- and time-related pattern.
Depression of the stem cells providing the protective mucosal cells lining the intestinal tract wall
results in a denuding of the gut surface. This gastrointestinal syndrome is seen in individuals who have
received doses to the gastrointestinal tract in excess of about 5 Gy. Leakage of blood from damaged blood
capillaries results in severe anaemia and ingress of intestinal bacteria through the damaged blood vessels
results in septicaemia. The haematopoietic syndrome will manifest itself concurrently at these higher
doses.
Landolt-Brnstein
New Series VIII/4

2-12

2 Biological effects of ionising radiation

[Ref. p. 2-34

Damage to endothelial cells lining the alveolar air sacs may result in acute inflammation of the lungs
(pneumonitis) at doses in the range 5 - 15 Gy. This may occur after radiotherapy or after the inhalation of
high specific activity radioactive particles. If the individual survives the pneumonitis, lung fibrosis may
later develop which can also be life-threatening through loss of lung function.
At higher whole-body doses (>15 Gy), generalised shock occurs affecting the brain and the
cardiovascular system. Coma and death develop rapidly thereafter.
The range of doses associated with death from these syndromes after acute exposure to low-LET
radiation is given in Table 2.2. The ranges are based upon human data, supplemented by knowledge of
the form of the dose-response relationship derived from animal experiments. No individual would be
expected to die after receiving absorbed doses below about 1 Gy. The dose range where half the exposed
population would be expected to die without medical treatment is 3 to 5 Gy. Death would be likely at
doses between about 6 Gy and 10 Gy, unless they receive treatment to prevent infection and bleeding.
Above about 10 Gy death is assumed at present to be inevitable, even after attempts to stimulate the bone
marrow or bone marrow transfusion from a suitable donor. These estimates of lethality do not take
account of any concurrent radiation-induced damage (e.g. skin burns), or existing debilitating diseases.
Table 2.2. Range of doses associated with acute radiation syndromes in adults exposed to low-LET
radiation.
Whole body
absorbed dose
1-6 Gy
5-15 Gy
>15 Gy

Principal effect contributing to death


Damage to bone marrowa
Damage to the gastrointestinal tract and lungsb
Damage to nervous system and shock to the cardiovascular
system

Time of death after


exposure [days]
30-60
10-20
1-5

a) Dose range considered to result in 50 % of an exposed population dying (LD50) 3-5 Gy.
b) Damage to vasculature and cell membranes especially at high doses is an important factor in causing death.

2.3.4 Deterministic effects following partial body irradiation


2.3.4.1 Tolerance doses in adults after radiotherapy
Extensive experience in the treatment of patients undergoing radiotherapy has provided data upon which
to determine the tolerability of healthy tissues and organs to radiation. Called the tolerance dose by
clinicians, it is defined as the amount of radiation received during conventional treatment below which
unacceptable effects do not occur in more than a few percent of patients within 5 years following
treatment. The tolerance doses for some adult tissues are shown in Table 2.3 (children are usually less
tolerant to exposure). It is evident that the gonads, lens of the eye and the bone marrow are the most
radiosensitive.
2.3.4.2 Threshold doses in radiological protection
The limitations of using data on tolerance doses to derive threshold doses for radiological protection
purposes need to be recognised. In contrast to the precise exposure conditions of radiotherapy, exposure
of workers to high doses of low-LET radiation is most likely to be non-uniform and resulting from mixed
radiations. The tolerance dose therefore can at best be used as a cautious approximation to a threshold
dose.

Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

2-13

Table 2.3. Tolerance doses for deterministic effects in adults after fractionated radiotherapy treatment
Organ
Effect
Tolerance dose [Gy]
Total bone marrow
Blood cell depletion
1-2
Ovary
Permanent sterilisation
2-6
Testis
Permanent sterilisationa
3-4
5-10
Eye
Cataractb
Kidney
Nephrosclerosis
23
Liver
Loss of function, ascites
35
Lung
Pneumonitisc
40
Heart
Pericarditis
40
Lymph nodes
Hypoplasia, fibrosis
35-45
Thyroid, pituitary
Hypoplasia
>45
Other organs
Hypoplasia, fibrosis
>45
a) A significant but reversible, depression of sperm count occurs after about 0.1 Gy brief exposure.
b) About 2 Gy after a brief exposure.
c) LD50 after brief exposure is about 10 Gy.

The threshold doses recommended by the ICRP for the most radiosensitive tissues and organs are
summarised in Table 2.4. Thus the threshold dose for temporary sterility in the male for a single absorbed
dose in the testes is about 0.15 Sv. Under conditions of prolonged exposure, however, the dose rate
threshold is about 0.4 Sv y1. The corresponding values for permanent sterility are about 3.5 Sv and
2 Sv y1. The threshold dose for permanent sterility in women for a single absorbed dose is in the range
from about 2.5 Sv. For protracted exposure, the dose rate threshold is about 0.2 Sv y1.
Clinically significant depression of the blood-forming process occurs above a single bone marrow
dose of about 0.5 Gy. The dose rate threshold for protracted exposure is about 0.4 Gy y1. The tolerance
dose for death is in the range of 6 to 7 Gy if the radiation is spread over 30 fractions in a period of 6
weeks. Table 2.5 summarises the principal syndromes associated with whole body exposure.
Table 2.4. Estimates of the thresholds for deterministic effects in adults recommended in radiological
protection [91I2].
Equivalent dose rate
Equivalent dose brief
Tissue and effect
exposure [Sv]
protracted exposure [Sv y1]
Testes
Temporary sterility
0.15
0.4a
Permanent sterility
3.5-6.0
2.0
Ovaries
Sterility
2.5-6.0
>0.2
Lens
Detectable opacities
0.5-2.0
>0.1
Visual impairment (cataract)
5.0c
>0.15
Bone marrow
Blood cell depletion
0.5
>0.4b
a)

This dose is higher because differentiating cells are more radiosensitive than the stem cells
so the latter can replenish the differentiating cells at an adequate rate.
b) Supported by evidence of effects after chronic radiation of Beagle dogs.
c) Range 2-10 Sv.

Landolt-Brnstein
New Series VIII/4

2-14

2 Biological effects of ionising radiation

[Ref. p. 2-34

Table 2.5. Summary of acute radiation syndrome


Syndromes
Intestinal
Cerebral

Bone marrow

Critical organ
Latent period
Syndrome threshold [Gy]
Death threshold [Gy]
Death occurring within
Cause of death

Bone marrow
2-3 weeks
1
2
3-8 weeks
Haemorrhage, infection

Prodromal vomiting
Symptoms

Brain
20 min
20
50
2 days
Cerebral oedema, heart
failure
Minutes
Tremors, cramps, loss of
coordination, lethargy,
impaired vision, coma

Treatment

Palliative

Prognosis

Hopeless

Small intestine
3-5 days
3
10
2 weeks
Sloughing of gut, shock

1 hour
A few hours
Loss of appetite, vomiting,
Fever, breathlessness, internal
diarrhoea with bleeding,
bleeding, depletion of bone
fever, electrolyte and fluid
marrow leading to low blood
balance
counts
Barrier nursing, fluid and electrolyte replacement, transfusions of
blood cells, bone marrow transplants
Very poor
Dose-dependent and influenced
by treatment

2.3.4.3 Skin irradiation


Based upon extensive experience in the use of fractionated X and gamma radiation in radiotherapy,
(typically, 20 to 30 fractions each of 2 to 6 Gy over several weeks), various degrees of skin damage can
be observed according to the area and depth of skin involved, the absorbed dose and the duration and
frequency of the exposure. The earliest observable change is a transient reddening within a few hours
after exposure to doses above about 2 Gy; due to increased capillary permeability. This is followed after
moderate doses (about 5 Gy) two to four weeks later by a persistent reddening (the main erythematous
reaction) and peeling of skin (dry desquamation). This is due to secondary inflammation resulting from
the death of basal (stem) cells of the epidermis. Hair loss also occurs.
At higher doses (about 20 Gy), blistering (moist desquamation) occurs after about four to six weeks
due to the inability of basal cells in the irradiated area to divide and for viable basal cells to migrate into
the area at a sufficiently rapid rate. It is the health effect to be avoided in both radiotherapy and radiation
protection practice. The threshold doses for moist desquamation depends upon the area irradiated and the
penetrating powers of the radiation.
Ulceration is the result of infection following moist desquamation and may occur after about 6 weeks.
Necrosis due to irreversible damage to the basal cells of the dermis and the underlying blood vessels
occurs within two to three weeks after doses of tens of Gy. Late effects developing months to years later
include changes in pigmentation; atrophy of the epidermis, sweat glands and sebaceous glands and hair
follicles; and fibrosis.
Quantifying the threshold doses for these effects is complicated in practice by the multiplicity of
targets at different critical cell depths, which makes it difficult to select a single depth at which to specify
the dose to the skin. The depths at which the most serious effects arise are estimated to be in the range of
300 - 500 m. However, a conservative approach for protection purposes is to use shallower depths (20 100 m, typically 70 m) for monitoring specifications.
To prevent moist desquamation, the dose must be reduced as the radiation field is increased. To
illustrate the importance of field size, the tolerance doses following a single treatment with orthovoltage
X-rays was found to be 20 Gy for an area of 6 4 cm, and 11 Gy for an area of 15 20 cm. Following
fractionated treatment, the tolerance doses were estimated to be about 50 Gy and 30 Gy respectively for
the two field sizes. From experimental studies, the estimated dose threshold following exposure of large
areas of skin is about 20 Gy; and no acute tissue breakdown was observed at a dose rate of 0.4 Gy h1
with total doses of about 100 Gy.
Accidental over-exposure of industrial radiographers is a cause for concern in radiation protection. In
normal practices, ICRP recommends a limit on effective dose of 20 mSv per year, averaged over 5 years
with the further provision that the effective dose should not exceed 50 mSv in a single year [91I2]. This
limitation is on effective dose and is assumed to be adequate to prevent deterministic effects. However, an
additional annual limit is recommended for localised exposures in order to prevent deterministic effects to
the skin. It is 500 mSv averaged over any 1 cm2 regardless of the area exposed.
Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

2-15

2.4 Radiation-induced cancer


2.4.1 Cancer development
The development of cancer is the major late effect resulting from exposure to radiation. Cancer is
generally understood to develop in a number of stages. That is, for malignancies to be expressed a series
of events must occur in cells and the rate at which they occur is thought to be reflected in the way cancers
appear in the population over the course of time.
The development of cancer in tissues is a complex, multi-stage process that can be sub-divided into
four phases: neoplastic initiation; neoplastic promotion; conversion and progression. The sub-divisions
are necessarily simplifications of the overall process which is, in any event, somewhat variable between
different tumour types. However, they do provide a basis from which to interpret the cellular and
molecular changes involved [93U6, 00U8].
Neoplastic initiation encompasses the essentially irreversible cellular damage, which although not
necessarily expressed immediately, provides the potential in cells for the development of cancer. There is
good evidence that this initiation process results from damage to DNA leading to gene mutations in single
target cells in tissues. The critical damage is likely to be coincident damage to both DNA strands (DNA
double strand breaks, Section 2.2.2). Although a proportion of such double strand damage will be
repaired, completely error free repair of such damage, even at low doses, is not expected. Neoplastic
promotion can be seen as a process whereby initiated cells receive an abnormal growth stimulus and
begin to proliferate in a semi-independent manner. Conversion of these pre-neoplastic cells to a form in
which they are committed to become fully malignant is a central feature of the process of neoplastic
development. Such changes are now believed to be driven by further gene mutations accumulating within
the expanding population of pre-neoplastic cells.
Once the potential for full malignancy has been established, the subsequent progression of the disease
may depend upon further cellular changes that allow invasion of adjacent normal tissues, the circulation
of neoplastic cells in the blood and lymphatic systems and the establishment of metastases (secondary
tumour growths) at other sites in the body. It is this invasive process that provides principally for the fatal
effects of most common human tumours. On this basis, a single mutational event in a critical gene in a
single target cell in vivo can create the potential for neoplastic development. Thus, a single radiation track
traversing the nucleus of an appropriate target cell has a finite probability, albeit very low, of generating
the specific damage to DNA that results in a tumour initiating mutation. These initiated cells can then
develop by multistage processes into an overt malignancy. As a consequence, at the level of DNA
damage, there is no basis for assuming that there is likely to be a dose threshold below which the risk of
tumour induction would be zero. For radiation protection purposes, a progressive increase in risk with
increasing dose, with no threshold, is therefore assumed [95C2]. Whilst such a multistage mechanism is
considered to be the cause of many human tumours there are likely to be some tumours that may arise in
tissues where there has been deterministic damage (fibrosis) for such tumour types a threshold dose may
need to be exceeded before the tumour will occur. There are many examples of such tumour types in
animals and the development of radiation-induced bone tumours in man may also require a threshold dose
to be exceeded [00U8].
Radiation appears to be capable of causing tumours in nearly all tissues of the body, although the
frequency of appearance following a unit dose may vary markedly from one tissue to another.
Information on the dose related frequency of tumour induction by radiation is gained through follow-up
of groups of persons exposed to radiation. The observed tumour frequency can then be compared with an
age and sex matched control group, not exposed to radiation, to determine the increase in frequency due
to the radiation exposure. Extensive follow-up studies have been carried out on groups of persons
exposed to either external radiation or to internally incorporated radionuclides.

Landolt-Brnstein
New Series VIII/4

2-16

2 Biological effects of ionising radiation

[Ref. p. 2-34

Tumours induced by radiation are in general indistinguishable from those occurring spontaneously
and since cancer is not uncommon (about one in five die as a result of it in Western Europe and North
America), the problem of determining a relatively small excess due to radiation exposure is difficult. In
general, large exposed populations are necessary to obtain statistically meaningful results.
The chief sources of information on the risks of radiation-induced cancer are the A-bomb survivors
exposed to whole-body irradiation in Hiroshima and Nagasaki, patients with ankylosing spondylitis and
other patients who were exposed to partial-body irradiation therapeutically, either from external radiation
or internally incorporated radionuclides, and various occupationally exposed populations, such as
uranium miners and radium-dial painters. Some quantitative information on thyroid cancers may also be
obtained following the Chernobyl accident. Increasingly information is becoming available from
epidemiological studies on groups of persons occupationally exposed to radiation. In general, however,
the radiation exposures in these populations is relatively low and there is limited power in the studies to
obtain quantitative estimates of risks of radiation-induced cancer. Reports by the United Nations
Scientific Committee on the Effects of Atomic Radiation (UNSCEAR) provide a comprehensive review
of the data available [94U7, 00U8].
In its 1990 recommendations, the ICRP re-assessed the epidemiological data and this resulted in an
increase in estimates of the lifetime risk of radiation-induced cancer. Partly, this arose as a result of
revised dosimetry for the A-bomb survivors and a longer follow-up of the population, but mainly it was
attributed to a change in the model now used to project lifetime risks [91I2]. Similar calculated values of
lifetime risk have been published by UNSCEAR [94U7, 00U8].

2.4.2 Dose-response relationships


2.4.2.1 Assessment of lifetime risk
There is always a minimum period of time between irradiation and the appearance of a radiation-induced
tumour. This period is termed the latent period and its length varies with age and from one tumour type to
another. Some types of leukaemia and bone cancer have latent periods of only a few years but many solid
tumours have latent periods of ten or more years. For leukaemia and bone cancer there is fairly good
evidence that the risk is almost completely expressed within about twenty-five years following exposure.
For solid tumours of longer latency, such as those of the GI tract , liver and lung it is not yet clear whether
the incidence of these tumours passes through a maximum and declines with time following exposure,
whether the risk levels out, or alternatively increases indefinitely during the remainder of life.
To project the overall cancer risk for an exposed population, it is therefore necessary to use models
that extrapolate over time data based on only a limited period of the lives of the individuals. Two such
projection models have generally been used:
(a) the additive (absolute) risk model which postulates that radiation will induce cancer independently of
the spontaneous rate after a period of latency, variations in risk may occur due to sex and age at
exposure as well as the tissue exposed.
(b) the relative (multiplicative) risk model in which the excess (after latency) is given by a constant (or
time-varying) factor applied to the age dependent incidence of natural cancers in the population.
In most cases the spontaneous risk of cancer increases with age and therefore the relative risk model
will predict an increasing incidence of radiation-induced cancer with age. This model also gives different
risks of radiation-induced cancer in different populations, depending on the national cancer incidence.
Data available from the A-bomb survivors in Japan and from studies on uranium miners suggest the
relative risk projection model gives a better fit to the data, at least for some of the most common cancer
types. Despite this there are indications from a number of exposed groups that the risk of cancer starts to
decline many years after exposure. This has been well documented for leukaemia, but has also been
Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

2-17

observed in the case of bone cancers (patients in Germany given 224Ra), thyroid cancers (US follow-up
study after thymus irradiation), solid cancers (patients treated for ankylosing spondylitics) and possibly
lung cancers in the uranium miners exposed to radon and its decay products [88U5; 00U8]. These results
suggest that for the Japanese population the excess risk may ultimately decrease with time and thus
relative risk projection models applied over a lifetime could result in an overestimate of the cancer risk.
2.4.2.2 Effects of dose and dose rate
The total radiation dose and the dose rate both influence cancer induction and are linked to the form of the
dose-response relationship. For radiological protection purposes tumour induction is generally assumed to
increase with increasing dose, with no threshold, as indicated above. However, studies using cells in
culture reveal that for many endpoints, including mutation, the dose-response for exposure to low-LET
radiation is not linear, but that the effectiveness of radiation, per unit dose, increases as the dose increases.
At very low doses, where there is a low probability of more than one radiation event occurring in a cell
nucleus it may be expected that the effect is linearly related to dose. At higher doses, where multiple
ionising events within a single cell are commonplace, damage arising from interactions between two or
more events becomes more probable. Ultimately, at high doses cell killing will progressively reduce the
risk of tumour induction. For single (acute) radiation exposures cell killing starts to become significant at
doses of a few gray. The generalised dose-response is given in Equation 3 (Section 2.2.3.2).
The difficulty in assessing risks of cancer following exposures to low-LET radiation at low doses and
dose rates is illustrated in Fig. 2.9. This gives, schematically, data points and possible dose-response
curves for cancer induction. Frequently, as in this example, information is only available at relatively high
doses. An approach commonly used in risk assessment is to fit a linear dose-response relationship to the
data (curve B) a procedure usually considered to give an upper limit to the risk at low doses. This will be
the case unless significant cell killing has occurred. If this linear relationship is due to single tracks acting
independently then the effect per unit dose would be expected to be independent of dose magnitude and
dose rate. In practice, however, this is not generally observed and the linear quadratic relationship (curve
A) frequently gives a better fit to the data at low to intermediate doses implying that at higher doses
damage is the result of both single and multiple tracks. At still higher doses cell killing becomes
significant with a consequent reduction in tumour yield.

Low LET

A - High absorbed doses and high dose rates


B - Linear, no threshold
C - Low dose rate
D - Limiting slope for low dose rate

Induced incidence of cancer

High LET

C
D

Absorbed dose D [Gy]

Landolt-Brnstein
New Series VIII/4

Fig. 2.9. Dose-response relationship for radiationinduced cancer: possible inferences are illustrated in
extrapolating data available at high doses and high dose
rates to response at low doses and dose rates for lowLET radiation; [based on 90N2].

2-18

2 Biological effects of ionising radiation

[Ref. p. 2-34

With a progressive lowering of the dose and the dose rate, allowing more opportunity for repair of
damage, curve C might be obtained. A point may ultimately be reached at which multiple track events
make a negligible contribution to tumour incidence and damage is produced only as a result of single
tracks acting alone giving a linear response (curve D) with the effect directly proportional to dose (slope
1, the risk coefficient). A similar response would be obtained by lowering the dose rate alone as even
with high total doses the rate of build up of lesions would be slower and the opportunity for multiple track
events would decrease. Hence in the limit, curve D, could be achieved either by reducing the dose to very
low values so that effects are independent of dose rate or by reducing the dose rate to very low values.
The approach used for assessing risks at low doses and low dose rates of low-LET radiation is described
in Sections 2.4.5 and 2.4.6. For high-LET radiation it is assumed that there is no dose rate effect and the
response is proportional to dose for doses below those at which there is cell killing.
The data on the A-bomb survivors provide information on risks of cancer in a range of tissues,
although to date no quantitative information is available for radiation-induced cancers of the liver, cells
on bone surfaces, thyroid and skin. Information on radiation-induced cancer in these tissues is, however,
available from other epidemiological studies summarised in Table 2.6. The principal studies used to
quantify the effects of both external radiation and internally incorporated radionuclides are summarised
below.
Table 2.6. Human populations available for risk estimation
Atomic bombs
Medical diagnosis
Medical therapy

Occupational exposure
Radiation accidents

Japanese survivorsa
Marshall islandersa,b
Multiple fluoroscopies (breast)a
Prenatal irradiationa
Thorotrast injectionsc
Pelvic radiotherapy (cervix)a
Spinal radiotherapy (ankylosing spondylitis)a
Neck and chest radiotherapy (thyroid)a,b
Scalp radiotherapya
Radium treatmentc
Uranium minersc
Radium ingestion (dial painters)c
Radiation workersa,b,c
Chernobyla,b

a) Exposure to external radiation


b) Internal exposure to / internal emitters
c) Internal exposure to emitters

2.4.3 Exposures to external radiation


2.4.3.1 The A-bomb survivors in Japan
The mortality experience of the Hiroshima and Nagasaki A-bomb survivors has been the single most
important source of information on the risk of radiation-induced cancer. This population has been the
subject of a comprehensive follow-up since 1950. Information is available on the exposure of individuals
to whole body radiation at a range of ages. Data on mortality from radiation-induced cancer that became
available in the 1980s on the population of more than 90,000 people in the Life Span Study (LSS)
necessitated a revision of previous risk estimates [87P3, 90S3]. There were a number of components to
this change. The first was a revision of the dosimetry (termed DS86) to allow, amongst other factors, for
the high humidity in the air over the cities which has substantially reduced the neutron dose at Hiroshima
from the earlier 1965 (T65) estimates which were based on measurements in the dry atmosphere of the
Nevada desert. Improved estimates were also made of the yield of the Hiroshima bomb (increased from
12.5 to 15 ktonnes), the shielding provided by buildings and of tissue and organ doses. The second was
Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

2-19

that the number of excess fatal cancers in the population increased due to the longer period of follow-up
(to 1985) and an estimate of the cancers occurring in the period 1945-1950 was made. The third, and most
significant change, was that relative risk, rather than additive risk models appeared to provide a better
basis for assessing lifetime risk of most solid cancers (Section 2.4.2.1).
UNSCEAR [88U5] in a report to the General Assembly provided the first information on
radiation-induced cancer risks for a number of tissues in the Japanese population based on relative risk
projection models. The total cancer risk in the population, at high dose and high dose rate, was then
estimated to be 7-11 102 Sv1 using age-averaged and age-specific constant relative risk models. This
compared with the Committee's 1977 assessment of 2.5 102 Sv1 [77U2] at high dose rate using the
additive model. Because children and young persons are more sensitive to radiation than adults. The
application of age specific risk coefficients therefore increases the predicted numbers of radiation-induced
cancers in the whole population compared with that for a working population (Section 2.4.6).
These risk estimates for whole body radiation exposure were based on an extrapolation into the future
which is somewhat uncertain for solid cancers because two-thirds of the Japanese survivors were still
alive and two-thirds of the cancer risk had still to be expressed. Up to 1985 about 80 excess leukaemias
and 260 excess solid cancers had occurred in the LSS population for whom DS86 doses were available
out of a total of about 6000 cancer deaths [87P3]. The risk of radiation-induced leukaemia is more certain
than that for solid cancers, however, as few more excess cases are now expected. There remain
uncertainties in extrapolating the cancer risks based on the Japanese population exposed to radiation at
high dose rates to the low doses and dose rates relevant for radiological protection purposes (see
Section 2.4.5).
In a more recent report on the LSS, Pierce et al [96P1] reported on five more years of follow-up
(1986-1990). Their analysis included an additional 10,500 survivors (86,572 in total). During 1950-1990
there have been 7827 cancer deaths, of which it is estimated there are 87 excess leukaemias and 334 solid
cancers. The mortality curve for all solid cancers combined shows essentially a linear dose-response in
the range 0-3 Sv, whereas for leukaemia the trend in dose is non-linear with an upward curvature. The
radiation-induced leukaemia risk seems to have been almost completely expressed during the follow-up
period, and the lifetime excess absolute risk of leukaemia associated with an acute dose of 1 Sv has been
estimated as being about 1 %. However, in contrast to leukaemia, nearly a quarter of the
radiation-induced solid cancers are estimated to have arisen in the most recent five-year period of the
mortality follow-up, i.e. 1986-90 [96P1]. Since most of the A-bomb survivors exposed at young ages are
still alive, the future pattern of cancer risks in this group will be important in determining lifetime risks. A
significant increase in the risk of solid cancers is now seen at doses down to about 50 mSv [00U8].
2.4.3.2 Thyroid cancer
A number of epidemiological studies provide information on cancer risks in individual tissues. Groups of
children and young persons who received thyroid irradiation, and who can be used to derive risk
coefficients for thyroid cancer, include children who received X-ray treatment for thymic enlargement,
patients treated in US hospitals for thyrotoxicosis and other benign lesions of the neck and patients who
received X-ray treatment for thyroid disease [85N1, 85S4, 00U8]. In the majority of cases, particularly in
the young, thyroid cancer is not fatal. The mortality from radiation-induced thyroid cancer is expected to
be about 10 % of the incidence. There is also evidence that the risk in adults is about half that in children
and that the risk in females is about twice that in males. For a population uniformly exposed to external
radiation the risk of fatal thyroid cancer is estimated to be 8.0 104 Sv1 assuming a 5 year latent period
[91I2]. In human populations given iodine-131 for non-therapeutic reasons, and who received doses well
below 2 Gy, no significant excess of thyroid cancers has been observed. This suggests a risk coefficient 3
to 4 times less than that obtained following external radiation at high dose rates [85N1]. Data on thyroid
cancer incidence in children in areas of the former Soviet Union that were contaminated with fall-out
from Chernobyl indicate an increased risk of thyroid cancer in some areas. To date the data are
insufficient to provide quantitative risk estimates [00U8]. Thyroid cancer risks from exposures to
radioiodine are considered further in Section 2.4.4.
Landolt-Brnstein
New Series VIII/4

2-20

2 Biological effects of ionising radiation

[Ref. p. 2-34

2.4.3.3 Skin cancer


An ICRP Task Group [91I3] reviewed data on the risks of skin cancer. Most of the data came from
groups given partial body irradiation in the course of medical treatment, although some data were also
available from occupationally exposed groups, in particular radiologists and radiation technicians and
uranium mining populations. Little information is available from the A-bomb survivors. On the basis of a
relative risk model, the Task Group calculated a risk of fatal skin cancer for exposure of a general
population of 2 104 Sv1 at low doses, on the assumption that 0.2 % of cases would be fatal. They
stressed the uncertainty in assessing the temporal pattern of radiation-induced skin cancers.
2.4.3.4 Breast cancer
Data are available on radiation-induced breast cancer from follow-up studies on the A-bomb survivors as
well as from studies of patients in North America given fluoroscopy examinations for tuberculosis or
treated for acute postpartum mastitis. Risks calculated from either population are little different, based on
additive projection models. ICRP has based its risk estimate of 2 103 Sv1, for a mixed population of
men and women, on the data on the A-bomb survivors. The risk of breast cancer also varies considerably
with age at exposure. Thus, for exposure in the first decade of life, the risk is about 4 times that at ages
40-50 years [93M4].

2.4.4. Exposure to internally incorporated radionuclides


Human data on cancer induction from internally incorporated radionuclides are available for only a few
radionuclides and have been reviewed by UNSCEAR [94U7, 00U8]. Quantitative data for risk estimation
are available only for alpha particle emitting radionuclides.
Limited data are available on humans exposed to / emitters. A number of epidemiological studies
have followed groups exposed to 131I. These studies cover a wide range of doses, varying from very high
doses delivered in the treatment of hypothyroidism to the low doses received by patients exposed to
diagnostic procedures or exposed to radiation from fallout in the Marshall Islands. The information
available provides little evidence that exposure to 131I is associated with a risk of thyroid cancer, although
in some cases the follow-up is relatively short. This lack of effect, compared with the effect of external
radiation, may be due to an effect of dose rate or to differences in the distribution of dose within the
gland. There may also be differences due to ages at exposure. As in the case of external radiation the
groups were predominantly young persons. The extent to which exposures to 131I has contributed to the
increased risk of thyroid cancer following the Chernobyl accident is still uncertain.
Some very sparse data on tumour induction are available on a few individuals given 32P, 35S and 59Fe
for medical reasons and there is some information on persons in the Southern Urals exposed to 90Sr who
used water from the Techa River for drinking and irrigation [94K1]. A number of studies have also
considered the effects of radionuclides in weapons fallout or in discharges to the environment from other
nuclear facilities. These data do not at present provide a basis for assessing risks from intakes of /
emitting radionuclides.
The available information on -particle emitters covers groups exposed to radium isotopes (224Ra,
226
Ra, 228Ra) where bone tumours are the predominant late effect, and Thorotrast (colloidal 232ThO2)
which principally results in irradiation of the liver, spleen and bone marrow, with tumours arising mainly
in the liver and bone marrow (leukaemia). Information is also available in man on lung cancer following
occupational exposure to radon and its decay products. A number of epidemiological studies of domestic
exposure to radon have been published and others are presently under way, to date the data are generally
consistent with risks obtained from worker studies although exposures are lower and have a reduced
sensitivity for obtaining quantitative risk estimates. Twenty-six men who worked with plutonium in North
America on the Manhattan project during the Second World War have also been studied (estimated body
Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

2-21

contents 52-3180 Bq). Seven individuals had died by 1991. The causes of death were lung cancer (2
cases), myocardial infarction, arteriosclerotic heart disease, accidental injury, respiratory failure due to
pneumonia/congestive heart failure and osteosarcoma of the sacrum. Three men also had a history of skin
cancer [91V3]. There is a high probability that the bone cancer was caused by exposure to plutonium as
the spontaneous risk is about 1 in 2000.
ICRP [91I2] has recommended the use of radiation weighting factors wR for calculating the equivalent
dose to tissues and thus interpolating between the effects of high and low-LET radiation. The wR for particle irradiation is taken to be 20.
2.4.4.1 Radium-226/228 luminisers
An increased incidence of bone cancer and of head sinus carcinoma has been observed in persons in the
USA exposed to long-lived radium, particularly in painters of luminous dials, but also radium chemists or
persons treated with radium salts for a possible therapeutic effect [86R2, 94R1]. These persons became
internally contaminated with pure 226Ra (t1/2 = 1,600 years) in some cases, and in other cases with various
mixtures of 226Ra and 228Ra (t1/2 = 5.8 years). Bone cancers and head sinus carcinomas have arisen in
these populations. The majority of these cancers had appeared by 1969, although three bone tumours have
appeared since then and more recently head cancers have appeared at a greater rate than bone cancers.
The radium isotopes deposit principally in the skeleton and the bone sarcomas appear to have been
induced by particles from either the 226Ra or 228Ra decay series. The head sinus carcinomas are thought
to be caused mainly by the accumulation of decay products of radon (222Rn) gas in the frontal sinuses and
mastoid air cells. This radon is produced by the decay of 226Ra in the bone.
Except for the bone sarcomas and head sinus carcinomas no definite excess in other types of
malignancy, including leukaemia, is presently ascribed to the internal deposition of long-lived radium.
The follow-up study on this population was essentially discontinued in the USA in the mid 1990s.
2.4.4.2 Radium-224 patients
The effects of intakes of radium has also been studied in German patients injected with 224Ra shortly after
World War II. The study group consists of a population of 682 adults and 218 juveniles (age at first
injection varied between 1 and 20 years) who received weekly or twice weekly intravenous injections of
224
Ra, mainly for the treatment of bone tuberculosis or ankylosing spondylitis [86M1, 94S5]. The last
bone tumour occurred in 1988, 41 years after the injection of 224Ra into a three-year-old boy and is the
only bone sarcoma reported in this series since 1974. Very few new tumours are now expected and
follow-up of the population is now limited.
Based on the information on the incidence of bone cancers following intakes of 224Ra and average
bone dose from its deposition in the skeleton, ICRP [91I2] has adopted a total risk estimate for fatal
cancer of 5 104 Sv1 (assuming a radiation weighting factor wR for -particle irradiation of 20).
2.4.4.3 Miners exposed to radon
An increased mortality from lung disease has been observed in under-ground miners working in
Czechoslovakia, Canada, United States of America and Sweden exposed to radon (222Rn) and its decay
products [88B3, 98B5].
The increase in mortality from lung cancer has been correlated with air concentrations of radon in
different mines and the duration of exposure. Bronchial stem cells and secretory cells in the airways are
considered to be the main target cell for the induction of lung cancer resulting from radon exposure.
There are many difficulties in calculating the radiation dose to these cells as a result of exposure to radon

Landolt-Brnstein
New Series VIII/4

2-22

2 Biological effects of ionising radiation

[Ref. p. 2-34

decay products (expressed in working level months1). The radiation dose over the working life must be
taken into account and the dust loading of the atmosphere known as it determines the extent of the uptake
of radon decay products onto the respirable particles. In addition to any possible synergistic effects
between smoking and radon exposure, the presence of dust, diesel fumes and other possible carcinogens
in the mine atmosphere causes some uncertainty as to whether an excess of cancer can be attributed to
radiation alone. The BEIR VI Committee [98B5] recommended two models for estimating radon risks
based on its analysis of the data on radon-exposed miners, without expressing a preference for either. One
of the BEIR VI models takes account of factors such as total exposure, age and average radon
concentration. Risks predicted under the latter model are about 50 % greater than those based on the
former model. The BEIR VI Committee also considered both multiplicative and submultiplicative
versions of these models. The risk predicted for smokers under the submultiplicative form of each model
is only slightly smaller than that based on the multiplicative version. In contrast, the risk for non-smokers
under the submultiplicative assumption is about twice that under the multiplicative version of the
corresponding model.
Based on the various combinations of the BEIR VI models, the lifetime risk of lung cancer for
smokers in the UK would lie in the range 10 %-15 %, while that for non-smokers would be in the range
1 %-3 %. For a general population of smokers and non-smokers, the range in lifetime risks would be
about 3 %-5 %. The BEIR VI model can also be used to calculate total risks of lung cancer in a
population in absolute terms. Thus in the UK population lung cancers attributable to the mean domestic
radon concentration of 20 Bq m3 would be in the range of 2000-3300 per year, based on the above
models. Taking into account the proportion of non-smokers in the population, it can be estimated that
about 500-1300 of radon-associated deaths would arise among non-smokers.
For a working population, ICRP [91I2] have adopted a risk factor for lung cancer of 0.68 102 Sv1
based on data from the the A-bomb survivors.
2.4.4.4 Thorotrast patients
Thorotrast is colloidal thorium dioxide. In the late 1920s it began to be injected into the arteries of
patients for use in diagnostic radiology as an X-ray contrast material. The average dose of about 25 ml of
Thorotrast contained 5 g of thorium with an activity (from -particles) of about 20 kBq 232Th with
additional radioactivity from its decay products. The colloidal Thorotrast was cleared from the
bloodstream by uptake into phagocytic cells depositing about 60 % in liver, 30 % in spleen and 10 % in
red marrow. Extensive epidemiological studies in Portugal, Sweden, Denmark, the United States, the
Federal Republic of Germany and Japan have shown that retention of thorium dioxide particles in the
liver and in the bone marrow resulted in an increased risk of liver tumours and leukaemias as well as liver
cirrhosis and other cardiovascular diseases [84V1, 94V2]. On the basis of an injected dose of 25 ml the
dose to the liver is estimated to be 0.25 Gy y1 (high-LET). Present estimates, based on a latent period of
20 years, suggest a lifetime risk of liver cancer following exposure to Thorotrast of about 0.15 102 Sv1
(assuming a wR for -particle irradiation of 20), about half this risk is expected to be expressed by 40
years after exposure [88B3, 91I2].

1 WL is any combination of the short-lived decay products of radon per litre of air which will result in the ultimate emission of 1.3
105 MeV of particle energy. A WLM results from exposure to a concentration of decay products in air of 1 WL for an average
working month of 170 hours.

Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

2-23

2.4.5 Dose and dose rate effectiveness factors (DDREFs)


Risk coefficients for radiation-induced cancer are based mainly on population groups exposed at high
doses and high dose rates as described above. Studies at the molecular, cellular, tissue and whole animal
level have demonstrated that radiation damage increases with dose and that, at least for low-LET
radiation, at high dose rates it is often greater per unit of exposure than at low dose rates. Thus, although
the assumption normally made for radiation protection purposes is that the dose-response curve for cancer
induction is linear, with the risk proportional to dose, in practice a dose and dose rate effectiveness factor
(DDREF) has commonly been used to allow for a reduced effectiveness of radiation in inducing cancer in
man at low doses and low dose rates. The choice of a suitable DDREF has caused considerable debate
with relevant data being available from cellular and animal studies, as well as human epidemiology.
ICRP in its 1990 recommendations based estimates of DDREF principally on an analysis by Pierce
and Vaeth [89P2] of the data from the Japanese survivors. This analysis shows that the data do not allow
for a reduction factor of much more than about 2. Other epidemiological data showed little evidence of
dose rate effects although studies on thyroid cancer incidence [85S4] and breast cancer mortality [89M3]
indicate possible reduction factors of up to 3 or 4. As a consequence ICRP adopted a DDREF of 2,
recognising that the choice is somewhat arbitrary and may be conservative. In practice, the DDREF
would be expected to vary with tissue and with exposure conditions although a single value had to be
assigned for protection purposes. A better understanding of the mechanisms involved will be essential for
improving understanding of the effects of both dose and dose rates on radiation-induced tumour induction
in man. A summary of values of DDREF recommended by national and international bodies is given in
Table 2.7. No DDREF is recommended for high-LET radiation (i.e. DDREF = 1).
Table 2.7. Summary of dose and dose rate effectiveness factors for radiation-induced cancer
Reference
DDREF
Source
ICRP 1977
77I1
2
NCRP 1980
90N2
2-10
UNSCEAR 1986
86U4
up to 5
UNSCEAR 1988
88U5
2-10
BEIR 1990
90B4
2
ICRP 1991
91I2
2
UNSCEAR 1993
93U6
<3
UNSCEAR 2000a
00U8
<3
a)

3 for hereditary disease

2.4.6 Risk coefficients for protection


In the last few years a number of reports have been published which have calculated risks of radiationinduced cancer for different populations. They have been based predominantly on information derived
from the A-bomb survivors but supplemented by data from other epidemiological studies as summarised
above. Most risks have been calculated for the general population, although a number of reports have also
given risks for workers. These tend to be lower (by about 20-40 %) because of the greater risk to children
and young persons calculated using the relative risk projection model for most solid cancers.
The assumption made for protection purposes is that the incidence of radiation-induced cancer
increases with the dose, with no threshold. Thus they are stochastic in nature. Tables 2.8 and 2.9
summarise the information on risks of radiation-induced cancer at high doses and high dose rates
published in recent years by UNSCEAR [88U5, 00U8], BEIR [90B4], NRPB [93M4] and ICRP [91I3],
using mainly relative risk projection models for most solid cancers. In the majority of studies lifetime
risks of cancer have been calculated, although NRPB also gave risks to 40 years after exposure (the then
period of follow-up of the A-bomb survivors). UNSCEAR [88U5] calculated risks based on both an ageLandolt-Brnstein
New Series VIII/4

2-24

2 Biological effects of ionising radiation

[Ref. p. 2-34

averaged and an age-specific constant relative risk models. BEIR V [90B4] calculated risks to a US
population and gave values for a number of tissues using time-varying relative risk models for some
cancers (leukaemia, respiratory tract, breast cancer in females). It is noteworthy that BEIR V, unlike
UNSCEAR, calculated excess cancer deaths, not early deaths. The former risk is about 20-25 % less than
the latter reflecting the baseline cancer rate in the population. ICRP [91I2] calculated risks for a 'world'
population based on an average value for five populations (Japan, UK, USA, Puerto Rico, China) and on
transferring both absolute and relative risks across populations.
Table 2.8. Estimated lifetime fatal cancer risks in populations (all ages, both sexes) associated with
exposure to low-LET radiation at high doses and high dose rates, based on a multiplicative projection
model
Source
UNSCEAR 1977
UNSCEAR 1988
BEIR V 1990
ICRP 1991
Muirhead 1993
a)
b)
c)
d)
e)

Reference

Population

77U2
88U5
90B4
91I2
93M4

Japan
USA
Five nations
UK

Fatal cancer risk


[102 Sv1]

2.5a
7-11b
7.9c
10.0d
4.9 - 11.80

additive model
range based on age-averaged and age-specific constant relative risks
see text (Section 2.4.6)
average value based on US, UK, Japan, Puerto Rico and Chinese populations. Risk for
workers 8.0 102 Sv1
risk calculated to 40 years after exposure and lifetime assuming age-specific relative
risks. Risk for workers 5.9-10.1 102 Sv1.

Note: These values are for acute doses only and do not include an adjustment for dose rate.

Table 2.9. Lifetime fatal cancer risks given by UNSCEAR 2000 [00U8].
Fatal cancer risk

Leukaemia
Solid Cancers
Males
Females

1 Sv

0.1 Sv

1%

0.05 %

9%
13 %

0.9 %
1.3 %

NOTE: These values are for acute doses only and do not include an adjustment for dose rate

Overall the lifetime risks calculated in recent years are not too different for the various studies, the
lowest value being for UNSCEAR [88U5] using age-averaged risk coefficients. ICRP [91I3] adopted a
rounded value of 10 102 Sv1 for the risk coefficient for fatal cancer at high doses and high dose rate
following exposure of a mixed population of all ages. Applying a DDREF of 2 gives a risk of 5 102
Sv1 for radiation protection purposes. Risk coefficients for individual tissues are given in Table 2.10. For
workers the risk coefficient adopted for radiation protection purposes is 4 102 Sv1. These risk
coefficients have been used by ICRP in developing the dose limits given in the 1990 recommendations
[91I3] and provide the basis for the International Basic Safety Standards [96I5] and the European Basic
Safety Standards [96E2].

Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

2-25

Table 2.10. Risk coefficients for fatal cancer adopted by ICRP


Organ or tissue
Bladder
Red bone marrow
Bone surface
Breast
Colon
Liver
Lung
Oesophagus
Ovary
Skin
Stomach
Thyroid
Remainder
Gonads
(hereditary disease)
Total

Fatal cancer risk coefficient [10-2 Sv-1]


ICRP 1991b
ICRP 1977a
Population
Workers

0.05
0.50
-

0.30
0.50
0.05
0.20
0.85
0.15
0.85
0.30
0.10
0.02
1.10
0.08
0.50
-

0.24
0.40
0.04
0.16
0.68
0.12
0.68
0.24
0.08
0.02
0.88
0.06
0.40
-

1.25

5.0

4.0

0.20
0.05
0.25
0.20

a) 77I1
b) 91I2

2.4.7 Low dose studies


The majority of studies on which risk estimates for radiation-induced cancer are based are for populations
exposed at high doses and high dose rates. Studies of low dose rate exposure generally involve low doses
and because of the likely low excess risks are hampered by lack of statistical power and possibly also by
confounding factors. However low dose rate studies can provide a check on the risks derived by
extrapolation from high dose rate studies. The main studies of interest are on workers who are
occupationally exposed although some data are also available on risks in children following exposures in
utero and on persons from areas of high natural background.
2.4.7.1 Occupational exposures
Several studies have been conducted of nuclear industry workers. In the USA, Gilbert et al, [89G1]
performed a joint analysis of data for about 36,000 workers at the Hanford site, Oak Ridge National
Laboratory and Rocky Flats weapons plant. Neither for the grouping of all cancers nor for leukaemia was
there an indication of an increasing trend in risk with dose.
In 1976 NRPB set up the National Registry for Radiation Workers (NRRW). The NRRW was
designed to investigate the effects of occupational exposures to ionising radiation by direct
epidemiological observations. The first analysis of the NRRW was published in l992 [92K2]. The main
findings were:

a strong healthy worker effect,


a statistically significant trend in leukaemia risk with dose, and
weaker evidence of a trend with dose for solid tumours.

Landolt-Brnstein
New Series VIII/4

2-26

2 Biological effects of ionising radiation

[Ref. p. 2-34

A healthy worker effect means that death rates in the group of workers studied are lower than in a
group of the general population of the same age and sex. This is a common finding in epidemiological
studies of working populations. The observation of a healthy worker effect in the NRRW cohort was
reassuring, but not unexpected. The more important findings were a trend with dose for leukaemia and a
(non-significant) trend for solid tumours. The risk factors in the first NRRW analysis were compatible
with those recommended by the ICRP but the confidence intervals were wide.
To obtain more precise information on the risks of radiation work NRPB carried out a second analysis
of the NRRW with a larger cohort and a longer period of follow-up [99M6]. A comparison of the main
features of the two studies is shown in the box.
Comparison of NRRW cohorts for the first and second analyses
First

Second

No. of Workers

95,217

124,743

Collective dose [man Sv]

3,198

3,810

Mean dose [mSv]

33.6

30.5

Person-years

1.2 million

2 million

Total deaths

6,660

12,972

The second NRRW analysis provided further information on mortality among UK radiation workers.
As in the first analysis, there was a strong healthy worker effect, with mortality from all causes and all
malignancies less than that expected from national rates. The 90 % confidence intervals for the trend in
cancer risk with external dose are tighter than before, and they now exclude values more than four times
those seen among the Japanese A-bomb survivors, although they are also generally consistent with no
raised risk. For leukaemia excluding chronic lymphatic leukaemia (CLL) there is evidence, of borderline
statistical significance, of an increasing risk with dose and, as with solid cancers, the data are consistent
with the A-bomb findings. Further analyses should provide more information on risks in relation to
occupational radiation exposure.
A combined analysis of mortality among 95,673 workers (85.4 % men) in the US, the UK and Canada
has been published [95C1]. The combination of the data from the various studies increases the overall
power to study associations between radiation and specific cancers. The combined analysis covered a
total of 2,124,526 person-years (PY) at risk and 15,825 deaths, 3,976 of which were due to cancer. As
with the NRRW, mortality from leukaemias, excluding CLL was significantly associated with cumulative
external radiation exposure. There was no evidence of an association between radiation dose and
mortality from all cancers. It was concluded that the results of the study did not suggest that current
radiation risk estimates for cancer at low levels of exposure are appreciably in error.
2.4.7.2 Background radiation
Studies of exposure to natural radiation (other than radon) have generally involved looking for any
geographical correlation with cancer rates. Such studies are difficult to interpret, however, owing to the
effect of confounding factors such as socio-demographic variables and other factors that vary
geographically.

Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

2-27

2.5 Hereditary disease


Radiation damage to the male and female germ cells, resulting in an increase in hereditary disease, is the
other main late effect resulting from exposure to ionising radiation. Only very limited data on genetic
damage are available from human populations and estimates of risk have to be derived mainly from
animal studies.

2.5.1 Categories of genetic damage


Inheritance is the process by which the genetic information carried by the DNA in the cell nucleus is
passed from one generation to the next. This is essentially an orderly process but mutations do arise
spontaneously giving a positive background level of genetic damage. Hereditary effects may occur by
changes arising in the base sequence in the DNA of a single gene leading to gene mutation, or by
rearrangement of collections of genes within and between chromosomes causing chromosomal
aberrations. Both may be produced by radiation.
There are three main types of gene mutation namely dominant, recessive and X-linked. Every
individual receives a set of genes from each parent. If dominant, a gene mutation in one set of genes but
not in the other can express itself in spite of its counterpart from the other parent being normal. A
recessive gene mutation cannot be expressed unless the genetic material from both parents carries the
same mutation. Females have two X chromosomes and males one X and one Y, the Y chromosome being
virtually inert apart from factors for maleness. An X-linked gene mutation can readily express itself in the
male whereas in the female X-linked mutations will not express themselves unless both X chromosomes
carry the same mutation.
The normal chromosome number in man is 46. Chromosomal mutations are due to alterations in
chromosome numbers or structure. If the number of chromosomes is increased or decreased in the
fertilised egg this produces such profound effects that, except in a few instances, death is likely to occur
soon after conception; if a child survives it is likely to have severe physical and/or mental defects. There
is at present no good evidence for radiation-induction of diseases of chromosomal origin [82U3, 88U5].
The genetics of some inherited diseases are more complicated because some relatively common
chronic diseases have a genetic element but additional factors such as environment play a part in their
expression. Changes in the mutation rate will also alter the incidence of these 'multifactorial' diseases.
Examples of various categories of hereditary diseases have been reviewed by UNSCEAR [77U2];
examples are given in Table 2.11.
Most live-born children with inherited chromosomal mutations exhibit mental and/or physical
abnormalities. There is little or no chance of sufferers who reach adulthood reproducing and so passing
these defects on to their children. These conditions are therefore maintained in the population by new
mutations arising either spontaneously or induced by an environmental insult such as radiation. Dominant
mutations show up in the first generation after exposure as do X-linked mutations in males and may occur
in subsequent generations if they do not prevent childbearing. Recessive mutations, however, tend to
occur in later generations. When assessing the risks of radiation it is therefore necessary to allow for
hereditary effects which may not appear for several generations.

Landolt-Brnstein
New Series VIII/4

2-28

2 Biological effects of ionising radiation

[Ref. p. 2-34

Table 2.11. Examples of hereditary diseases [77U2].


Dominant disorders
Congenital cataract
Cystic kidney disease
Huntingtons chorea (progressive mental
retardation)
X-linked diseases
Haemophilia
Albinism
Colour blindness
Heart valve defects
Autosomal recessive diseases
Cretinism
Disorders of amino acid metabolism
Aplastic anaemia
Muscular dystrophy
Multifactorial diseases
Ankylosing spondylitis
Varicose veins
Cleft palate
Diabetes mellitus
Schizophrenia
Asthma
Chromosome anomalies
Downs syndrome

2.5.2 Risk coefficients for hereditary disease


So far no hereditary effects at levels that are statistically significant have been observed in human
populations exposed to radiation [88U5]. Neel et al [89N4] have reviewed all the genetic studies in
Hiroshima and Nagasaki on the children born to irradiated survivors. The 'end points' considered were
congenital defect, survival of liveborn infants, sex-chromosome aneuploidy and balanced chromosomal
exchanges, cancer with onset below age 20, mutations altering protein charge or activity, sex ratio, and
physical growth and development. The average conjoint parental gonad exposures for the parents was
about 0.5 Gy (based on DS86 dosimetry), the exact figure depending on the radiation histories of the
parents whose children formed the basis for a specific end point. No statistically significant effects were
observed. Taken together, the data suggest a lower limit for the doubling dose for genetic damage
following acute irradiation of approximately 1.4-1.8 Sv. This compares with a value of 0.3 Sv in the
mouse for acute exposure and 1 Sv for chronic exposure. The assumption made in calculating risks for
radiation protection purposes is that the incidence of radiation-induced hereditary disease increases with
the dose, with no threshold. Thus they are stochastic in nature.
In the absence of direct quantitative human data, animal studies have been used by UNSCEAR [88U5]
to assess the risk of radiation-induced hereditary disease in human populations. Tables 2.12 and 2.13
gives some estimates of the incidence of hereditary diseases in a population recommended by UNSCEAR
and ICRP in recent years. They are based on a doubling dose for hereditary disease of 1 Gy derived from
animal studies. In the most recent report by UNSCEAR [01U9] the risk of radiation-induced hereditary
disease has been appreciably reduced compared with its previous advice.

Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

2-29

Table 2.12. Incidence of genetic disease at equilibrium from parental exposure.


Incidence (low-LET)a [102 Gy1]
Disease classification
Chromosomal anomalies
Dominant and X-linked
Recessive
Multifactorial
Total

UNSCEAR,
1977
[77U2]

0.4
1.0
0.45
1.85

ICRP,
1977
[77I1]

UNSCEAR,
1982
[82U3]

UNSCEAR,
1988
[88U5]

ICRP, 1991
[91I2]

2.0

0.04
1.0
0.45
1.49

0.04
1.0
0.15
1.19

0.04
1.0
0.15
3.5b
2.4

a) Assuming doubling dose of 1 Gy.


b) Severity less than other genetic diseases, weighted by factor of 1/3.

Table 2.13. Incidence of genetic disease from one-generation exposure to low-LET, low dose rate or
chronic radiation [01U9].
Disease class
Chromosomal anomalies
Dominant and X-linked
Recessive
Multifactional
Chronic multifactional
Congenital anomalies
Total
a)

Baseline frequency
per 106 live births
4,000
16,500
7,500

Incidence (low-LET)a [102 Gy1]


1st generation
2nd generation
0.075-0.15
0.05-0.10
-

650,000
50,000
738,000

~0.025-0.12
~0.2
~0.3-0.47

~0.025-0.12
~0.04-0.10
0.11-0.32

Assuming doubling dose of 1 Gy

In its most recent recommendations ICRP [91I2] gives a risk of hereditary disorders of 2.4 102 Sv1
expressed over all generations following exposure of either parent (Table 2.12). This risk factor includes a
risk factor for multifactorial diseases. At present the information on such diseases is very limited and only
a tentative value is available. ICRP [91I2] have assessed the risk as 3.5 102 Sv1 following exposure of
either parent. The severity of multifactorial diseases are considered to be not as great as other hereditary
diseases so they are weighted by a factor of three, giving a risk factor of 1.2 102 Sv1. The genetically
significant exposure in a population will be less than this because a proportion of the population are older
than child bearing age. If the mean age of child bearing is 30 years and average life expectancy is 75
years then the probability of genetic harm resulting from exposure of the entire population is 30/75
(= 0.4) 2.4 102 Sv1 102 Sv1 [91I2] (Table 2.14).
For a working population the reproductive fraction is less than the entire population. For a working
population, the reproductive fraction is (30-18)/(65-18) = 0.25. The risk factor for workers is thus about
0.6 of that for an entire population (0.25/0.4) giving a risk factor for workers of 0.6 102 Sv1 (60 % of
1 102 Sv1) (Table 2.14).

2.6 Irradiation in utero


For the developing embryo and fetus there is evidence that deterministic effects, severe mental retardation
and cancer induction may occur following irradiation in utero. The risk of hereditary disease may be
taken to be the same as after birth.

Landolt-Brnstein
New Series VIII/4

2-30

2 Biological effects of ionising radiation

[Ref. p. 2-34

2.6.1 Deterministic effects


Evidence of the deterministic effects of radiation on the embryo and fetus is derived almost entirely from
animal experiments. It is necessary to extrapolate the results of these studies to predict the consequences
of radiation exposure in man.
The effects of radiation on the embryo depend on the time of exposure relative to its development.
When the number of cells in the embryo is small (i.e., in the first few days after fertilisation) and they are
not yet specialised, damage is frequently seen in animals as failure of the conceptus to implant or loss of
embryos, which would be seen in humans as miscarriage. However, recent evidence from in vitro human
embryo research has shown that the survival of even one cell in the early embryo before implantation can
allow normal development of cells to occur, since all the necessary genetic components are present in
each cell of the embryo at this early stage of development. The consequences of any of these cells
carrying a point mutation are unquantified, but the possibility of stochastic effects occurring cannot be
dismissed.
Malformations have been observed in rodent embryos at a stage when organs such as the brain,
skeleton, eyes and heart are developing. Congenital abnormalities are commonly found in the offspring of
rodents but any attempts to project the results to predict effects in man are fraught with difficulties.
With this cautionary note and bearing in mind, proposed human threshold doses for radiological
protection purposes for low-LET radiation: 0.05 Gy for reabsorption of pre-implantation embryos;
0.05 Gy for minor skeletal abnormalities; 0.2 Gy for functional disorders of the central nervous system;
and between 0.2 and 0.5 Gy for serious skeletal abnormalities and growth retardation, such information
provides a basis for guidelines to ensure that pregnant women are adequately protected [03I1]. Protraction
of the dose will reduce any effect.

2.6.2 Brain function


The human brain is probably the most complex organ in the body and its proper development and
function depend upon an elaborate sequence of events which must be coordinated temporally and
spatially. Any disturbance of this sequence could lead to abnormality since the normal function of the
nervous system depends upon the proper location of the neuronal cells.
A study of about 1600 children exposed in utero at Hiroshima and Nagasaki to various radiation doses
and at various developmental stages has shown about 30 cases of clinically severe mental retardation with
a greater incidence than expected in the higher dose groups. Excess mental retardation was not observed
following exposure up to 8 weeks from conception, was at a maximum between 8 and 15 weeks and then
was somewhat lower between 16 and 25 weeks. No effect was observed following exposures later than 25
weeks [84O2; 88O3].
The period of maximum sensitivity (8-15 weeks) corresponds with the timing of both of the major
waves of neuronal proliferation and migration within the cerebral cortex. Although the number of cases is
small, the data indicate an excess probability of 40% at 1 Sv received during the 8-15 weeks after
conception. The current results of IQ tests amongst those children exposed in utero indicates a general
downward shift in the distribution of IQ with increasing dose. A coefficient of about 30 IQ points Sv-1
relates to in utero exposure between 8-15 weeks after conception. A smaller shift is identified in the
16-25 week period [88S1].
This downward shift in IQ of 30 points Sv1 shown schematically in Figure 2.10 is consistent with the
observation of an incidence of 0.4 for a dose of 1 Sv. At doses of the order of 0.05 Sv, no effect would be
detectable in the general distribution of IQ, but at somewhat larger doses the effect might be sufficient to
show an increase in the numbers classified as seriously mentally retarded. The net result is that the end
point of serious mental retardation would appear to demonstrate a threshold, which is reasonably
consistent. The ICRP now believes that the phenomenon is deterministic with a threshold related to the
minimum shift in IQ that can be measured. It is not therefore included in the definition of radiation
detriment used for protection purposes.

Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

f
x

Retarded fraction f

( X )

2-31

Fig. 2.10. The shift to the left from (x) by x (30 IQ


points) increases the background retarded fraction f by
f. xm denotes the number of standard deviations below
IQ 100 to classify an individual as mentally retarded
[91I2].

x=0
(IQ 100)

2.6.3 Risk coefficients for cancer


Information on the risk of cancer following irradiation in utero has been reviewed by UNSCEAR [72U1,
77U2] and by the BEIR-III Committee [80B2]. Current risk estimates for radiation-induced childhood
cancer are based mainly on data collected in the Oxford Survey of Childhood Cancers (OSCC)
concerning obstetric radiography [75B6]. This study contains information on over 150,000 childhood
cancer deaths in Great Britain during 1953-81 and the same number of matched control children [87K3].
The OSCC, in common with other, smaller, case-control studies, indicates a relative risk of about 1.4
(40 % increase in risk) for childhood cancer associated with prenatal irradiation [89B7]. Concerns about
possible bias and confounding in these case-control studies have been raised for example, by Boice and
Miller [99B8] in view of issues such as the lack of evidence for a raised risk from cohort studies, and
the similarity of the relative risks for leukaemia and other cancers in the OSCC. In their review of these
issues, Doll and Wakeford [97D2] concluded that there is strong evidence against bias and confounding
as alternative explanations for the raised risks seen in the OSCC and other case-control studies. The doses
received by the fetus are uncertain; based on estimated average doses of about 10-20 mGy. Based on data
from the OSCC and information from UNSCEAR [72U1] on doses received in utero from obstetric
radiography the number of excess cancer cases (to 15 years of age) following irradiation in utero is
calculated to be about 6 102 Gy-1 [93M4]. Since slightly less than 50 % of childhood cancers consist of
leukaemia and other lymphatic/haematopoietic cancers [81O1] and the relative risks are similar for these
and other cancers, a risk of 2.5 102 Gy1 is calculated for leukaemia and 3.5 102 Gy1 for solid
cancers. As approximately half of all childhood cancers are fatal [81O1], the number of excess cancer
deaths is calculated to be 3 102 Gy1 (low-LET), comprising 1.25 102 Gy1 for leukaemias and 1.75
102 Gy1 for solid cancers. These risks are derived principally from follow-up studies on children
irradiated in utero with radiation doses up to a maximum of 10-20 mGy (low-LET). They are therefore
applicable for estimating risks at low doses and dose rates. There is also likely to be an additional risk of
cancer that will appear late in life but the information is very limited. In addition, follow-up of persons
exposed to A-bomb radiation in utero in Hiroshima and Nagasaki indicates that the raised cancer risk
continues into adulthood, although quantification of this risk is difficult [88Y1, 97D1].

2.6.4 Hereditary disease


Hereditary disease is considered in Section 2.5. Genetic studies in the offspring of atomic bomb survivors
have not shown any significant radiation-related increases in any measure of genetic damage employed.
In experiments in mice the sensitivity of fetal gonads was comparable to that of adult gonads or a little
lower [74S2]. It is therefore assumed that the risks of hereditary disease from in utero irradiation are the
same as after birth (2.4 102 Sv1) following exposure of either male or female germ cells. It may be the
risk will be lower in early embryogenesis and fetogenesis prior to the establishment of germinal tissues.

Landolt-Brnstein
New Series VIII/4

2-32

2 Biological effects of ionising radiation

[Ref. p. 2-34

2.7 Summary of risk factors for cancer and hereditary disease


The ICRP, in its most recent 1990 recommendations [91I2], considers four components of the detriment
(health effects) due to irradiation of the tissues and organs of the body at low doses when assessing the
overall effects of radiation. These include the probability of fatal cancer; the probability of non-fatal
cancer and the probability of severe hereditary disease, both weighted for severity relative to fatal cancer;
and the time scale of appearance of these detrimental effects. The risk factors developed by ICRP for
protection purposes are summarised in Table 2.14. The overall weighted severity values assigned to the
non-fatal cancers and severe hereditary diseases (including multifactorial diseases) each amount to about
one-fifth of the detriment associated with fatal cancer. In summary the aggregated detriment amounts to
7.3 102 Sv1 for a nominal population. It is somewhat less (5.6 102 Sv1) for a population aged 18-64
years who are occupationally exposed, when account is taken of the omission of younger persons who are
more radio-sensitive and the shorter mean potential period of reproduction. The temporal pattern of fatal
cancer risk is such that the period of maximum risk occurs in the seventh and eighth decades of life if the
multiplicative projection model is used to calculate the lifetime expression of the cancers in persons
exposed continuously to small annual doses at or below the dose limits.
Table 2.14. Risk factors for protection [102 Sv1] [77I1, 91I2]
ICRP 1991
ICRP 1977
Public
Workers
Fatal cancer
1.25
5.0
4.0
Hereditary defects
0.4a
1.0b
0.6b
Total
1.65
6.0
4.6
Total (weighted)c
7.3
5.6
a) Two generations
b) All generations
c) To allow for non-fatal cancers and years of life lost for cancers and hereditary disease.

2.8 Conclusions
Deterministic effects in tissues and organs are the result of the loss of substantial numbers of stem cells,
thereby cutting off the supply of functional cells. The consequence can be a temporary or permanent loss
of tissue function which may be life threatening. A characteristic of the dose-response relationship for
deterministic effects is that they are avoidable below a dose threshold. This is a reflection of sufficient
numbers of stem cells maintaining functional cell populations. Knowledge of dose thresholds has been
derived from the tolerance doses observed in radiotherapy. The tolerance doses vary with the tissue - the
gonads, the bone marrow, the gastrointestinal and the lens of the eye being the most sensitive. It is the
opinion of the ICRP that deterministic effects can be avoided if the presently recommended effective dose
limits (based upon limiting stochastic effects) and the annual equivalent doses for the lens of the eye and
the skin are not exceeded.
There are a number of important questions that remain to be answered in the assessment of the risks of
radiation-induced cancer in human populations. Very limited information is available at the low doses and
low dose rates that are important for radiation protection purposes and the risks have to be assessed from
populations exposed at high doses and dose rates by applying an appropriate dose and dose rate
effectiveness factor. Increasingly, however, epidemiological studies on groups of workers in the nuclear
industry are providing information on exposures at low doses and dose rates although at present any
estimates of risk have large uncertainties associated with them. With the development of these national
studies and by pooling them internationally these uncertainties should be progressively reduced.
Landolt-Brnstein
New Series VIII/4

Ref. p. 2-34]

2 Biological effects of ionising radiation

2-33

Continued follow-up of exposed populations, in particular the A-bomb survivors in Japan is needed for
validating current lifetime projection models. It seems unlikely that epidemiological studies will be able
to answer all the questions concerned with the effects of dose, dose rate, radiation quality and individual
sensitivity to cancer induction. Ultimately this must depend on a much better understanding of the
response of tissues to radiation. This will come increasingly from cellular and molecular studies designed
to understand the fundamental mechanisms involved in cancer induction.
The assumption made for protection purposes is that the incidence of radiation-induced cancer and
hereditary disease increases with the dose, with no threshold. Thus it is not possible to completely prevent
any risks. Protection standards must, therefore, be set to keep any risk to an acceptable level.
Table 2.15 gives a chronology of the first century of radiation protection.
Table 2.15. Chronology of the first century of radiation protection
Year
Event
1895
Discovery of X-rays (November 8)
1896
X-ray report made public (January 3)
Discovery of radioactivity (February)
First reports of possible X-ray injury;
Damage to eyes (March 3)
Skin effects first noted (April 18)
1901
X-ray lethality to mammals demonstrated experimentally
1904
First death in X-ray pioneer attributed to cumulative overexposure
(October)
1906
Law of radiosensitivity of tissues put forth
1911
1915
1920
1921
1925
1927
1928
1929
1931
1932
1934
1936
1941
1944
1950
1991

International radium standard and Curie unit


British Roentgen Society adopts radiation protection recommendations
First Standing X-ray Protection Committee
British X-ray and Radium Protection Committee issues first
memorandum
First tolerance dose proposed
Genetic effects of X-rays shown (drosophila)
Roentgen unit formally adopted, International X-ray and Radium
Protection Committee formed (forerunner of ICRP)
US Advisory Committee on X-ray and Radium Protection formed
(forerunner of NCRP)
USACXRP publishes first recommendations - 0.2 R/day
Concept of greater permissible dose for partial body irradiation (hands)
introduced
Discovery of neutrons
ICXRP recommends permissible dose of 0.2 R/day
USACXRP recommends reduction in permissible dose to 0.1 R/day
USACXRP recommends adoption of maximum body burden of 0.1 Ci
for radium
Suggested maximum permissible dose of 0.02 R/day
Maximum permissible concentration for inhaled radioactivity introduced
Rem and Rep introduced
ICRP set up
Publication of 1990 Recommendations of ICRP

Investigator
Roentgen
Becquerel
Edison
Morton
Stevens
Rollins
Dally
Bergonie
Tribondeau
Curie
ARRS
Mutscheller
Mller

Failla
Chadwick

Taylor
Parker
Parker

Please Note: Work on preparation of this Chapter was completed in March 2002. Readers should note
that ICRP is planning to issue new recommendations on radiological protection in 2005. This will take
into account more recent information on health effects published since its 1990 recommendations [91I2].

Landolt-Brnstein
New Series VIII/4

2-34

2 Biological effects of ionising radiation

2.9 References
72U1
74S2
75B6
77I1
77U2
80B2
81O1
82U3
84O2
84V1
85N1
85S4
86B1
86M1
86R2

86U4
87K3
87P3
88B3
88O3
88S1
88U5
88W1
88Y1

UNSCEAR: Ionizing Radiation : Levels and effects. 1972 Report to the general assembly, with
annexes. Vol. II: Effects. New York: United Nations, 1972.
Searle, A.G.: Adv. Radiat. Biol. 4 (1974) 121.
Bithell, J.F., Stewart, A.M.: Br. J. Cancer 35 (1975) 271.
ICRP.: Recommendations of the international commission on radiological protection. ICRP
Publication 26. Oxford: Pergamon Press, 1977.
UNSCEAR: Sources and effects of ionising radiation. 1977 report to the general assembly, with
annexes, 1977.
BEIR III.: Committee on the biological effects of ionising radiations. The effects on populations
of exposure to low levels of ionizing radiation. National Academy of Sciences. Washington DC:
National Academy Press, 1980.
OPCS. Cancer statistics: Incidence, survival and mortality in England and Wales. Studies on
medical and population subjects, No. 43, London: HMSO, 1981.
UNSCEAR: Ionizing radiation sources and biological effects. Report to the general assembly,
with annexes, 1982.
Otake, M., Schull, W.J.: Br. J. Radiol. 57 (1984) 409.
van Kaick, G., Muth, H., Kaul, A.: The German thorotrast study. Report No. EUR 9504 EN.
Luxembourg CEC (quoted in BEIR IV), 1984.
National council on radiation protection and measurements, NCRP, Report No. 80. Induction of
thyroid cancer by ionising radiation, 1985.
Shore, R.E.: J. Natl. Cancer Inst. 74 (1985) 1177.
Barendsen, G.W.: Responses of cultured cells, tumours and normal tissues to radiations of
different linear energy transfer. IN: Current Topics in Radiation Research, Vol. 4, pp 293-356
(Ebert and Howard, eds). North-Holland Publishing Company, Amsterdam.
Mays, C.W., Spiess, H., Chemelevsky, D., Kellerer, A.; in: Proc. Symp. "The Radiobiology of
Radium and Thorotrast", Gssner, W., Gerber, G.B., Hagen, J., Luz, A. (eds.), Neuherberg, 2931 Oct, 1984. Strahlentherapie 80, Suppl. 14-21 (1986) 27.
Rundo, J., Keane, A.T., Lucas, H.F.: Current (1984) status of the study of 226Ra and 228Ra in
humans at the Center for Human Biology; in: Proc. Symp. "The Radiobiology of Radium and
Thorotrast", Gssner, W., Gerber, G.B., Hagen, U., Luz, A. (eds.), Neuherberg 29-31 Oct, 1984;
Strahlentherapie 80 Suppl. 14-21 (1986).
UNSCEAR. Genetic and Somatic Effects of Ionizing Radiation. 1986 Report to the General
Assembly, with annexes (1986).
Knox, E.G., Stewart, A.M., Kneale, G.W., Gilman, E.A.: J. Soc. Radiol. Prot. 7 (1987) 177.
Preston, D.L., Pierce, D.A.: The effects of changes in dosimetry on cancer mortality risk
estimates in the atomic bomb survivors. Hiroshima, Radiation Effects Research Foundation,
RERF TR 0-87, 1987.
BEIR IV.: Health risks of radon and other internally deposited alpha-emitters. Washington DC:
National Academy Press, 1988.
Otake, M., Yoshimaru, H., Schull, W.J.: Severe mental retardation among prenatally exposed
survivors of the atomic bombing of Hiroshima and Nagasaki: A comparison of the T65DR and DS86
dosimetry systems. Hiroshima, Radiation Effects Research Foundation, RERF TR16-87, 1988.
Schull, W.J., Otake, M., Yoshimaru, H.: Effect on intelligence test score of prenatal exposure to
ionising radiation in Hiroshima and Nagasaki: A comparison of the T65DR and DS86 dosimetry
systems. Hiroshima, Radiation Effects Research Foundation, RERF TR3-88, 1988.
UNSCEAR: Sources, effects and risks of ionizing radiation. 1988 Report to the general
assembly, with annexes, 1988.
Ward, F.: Prog. Nucleic Acid Res. Mol. Biol. 35 (1988) 95.
Yoshimoto, Y., Kato, H., Schull, W.J.: Lancet 2 (1988) 665.

Landolt-Brnstein
New Series VIII/4

2 Biological effects of ionising radiation


89B7
89E1
89G1
89K4
89M3
89M5
89N4

89P2
90B4
90N2
90N3
90S3
91I2
91I3
91V3
92K2
93I4
93M4
93U6
94G2
94K1
94R1
94S5
94U7

2-35

Bithell, J.F., Epidemiological studies of children irradiation in utero. J.F. Bithell (1989), in: Low
dose radiation: biological bases of risk assessment, Baverstock, K.F., Stather, J.W. (eds),
London: Taylor and Francis, 1989.
Edwards, A.A., Lloyd, D.C., Prosser, J.S.: Chromosome aberrations in human lymphocytes - a
radiobiological review; in: Low dose radiation: biological bases of risk assessment, Baverstock,
K.F., Stather, J.W. (eds),. London: Taylor and Francis, 1989, p. 423.
Gilbert, E.S., Fry, S.A., Wiggs, L.D., Voelz, G.L., Cragle, D.L., Petersen, G.R.: Radiat. Res. 120
(1989) 19.
Kronenberg, A., Little, J.B.: Mutagenic properties of low doses of X-rays, fast neutrons and
selected heavy ions in human cells; in: Low dose radiation: Biological bases of risk assessment,
Baverstock, K.F., Stather, J.W. (eds), London: Taylor and Francis, 1989, p. 423.
Miller, A.B., Howe, G.R., Sherman, G.J., Lindsay, J.P., Yaffe, M.J., Dinner, P.J., Risch, H.A.,
Preston, D.L.: N. Engl. J. Med. 321 (1989) 1285.
Muirhead, C.R., Kneale, G.W.: J. Radiol. Prot. 9 (1989) 209.
Neel, J.V., Schull, W.J., Awa, A.A., Satoh, C., Otake, M., Kato, H., Yoshimoto, Y.: The genetic
effects of the atomic bombs: Problems in extrapolating from somatic cell findings to risk for
children. Low dose radiation: Biological bases of risk assessment, Bayerstock, K.F., Stather,
J.W. (eds.), London: Taylor and Francis, 1989, p. 42.
Pierce, D.A., Vaeth, M.: Cancer risk estimation from the A-bomb survivors: Extrapolation to low
doses, use of relative risk models and other uncertainties; in: Low dose radiation biological bases
of risk assessment, Baverstock, K.F., Stather, J.W. (eds.), London: Taylor and Francis, 1989, p 54.
BEIR V.: Health effects of exposure to low levels of ionizing radiation. Washington DC:
National Academy Press, 1990.
NCRP.: Influence of dose and its distribution in time on dose-response relationships for lowLET radiation. Washington DC: NCRP Report No. 64, 1980.
NCRP.: The relative biological effectiveness of radiations of different quality. NCRP Report No.
104, 1990.
Shimizu, Y., Kato, H., Schull, W.J.: Radiat. Res. 121 (1990) 120.
ICRP.: 1990 Recommendations of the international commission on radiological protection.
ICRP Publication 60. Annals of the ICRP 21, No.1-3. Oxford: Pergamon Press, 1991.
ICRP.: The biological basis for dose limitation in the skin. ICRP Publication 59. Annals of the
ICRP 22, No. 2. Oxford: Pergamon Press, 1991.
Voelz, G.L., Lawrence, J.N.P.: Health Phys. 61 (1991) 181.
Kendall, G.M., Muirhead, C.R., MacGibbon, B.H., OHagan, J.A., Conquest, A.J., Goodill,
A.A., Butland, B.K., Fell, T.P., Jackson, D.A., Webb, M.A., Haylock, R.G.E., Thomas, J.M.,
Silk, T.J.: Br. Med. J. 304 (1992) 220.
ICRU.: Quantities and units in radiation protection dosimetry. Publication 51, 1993.
Muirhead, C.R., Cox, R., Stather, J.W., MacGibbon, B.H., Edwards, A.A., Haylock, R.G.E.:
Docs. NRPB 4(4) (1993) 15-157.
UNSCEAR: Sources, effects and risks of ionizing radiation. 1993 Report to the general
assembly, with annexes, 1993.
Goodhead, D.T.: Int. J. Radiat. Biol. 65 (1) (1994) 7-17.
Kellerer, A.M., Burkhart, W. (eds.): Radiation exposure in the Southern Urals. The Science of
the Total Environ. Special Issue, 1994.
Rowland, R.E.: Radium in Humans. A Review of US Studies. ANL/ER-3 UC-408 Argonne
National Laboratory, 1994.
Spiess, H.: The Ra-224 Study: Past, Present and Future; in: Proc. Int. Seminar on Health Effects
of Internally Deposited Radionuclides: Emphasis on Radium and Thorium, Heidelberg,
Germany, 1994.
UNSCEAR: Sources, effects and risks of ionizing radiation. 1994 Report to the general
assembly, with annexes, 1994.

Landolt-Brnstein
New Series VIII/4

2-36
94V2

95C1
95C2
96E2
96I5
96P1
97D1
97D2
98B5
98M2
99B8
99M6
00U8
01U9
03I1

2 Biological effects of ionising radiation


van Kaick, G., Wesch, H., Luehrs, H., Liebermann, D., Kaul, A.: Epidemiological results and
dosimetric calculations - an update of the German thorotrast study; in: Proc. Int. Seminar on
Health Effects of Internally Deposited Radionuclides: Emphasis on Radium and Thorium,
Heidelberg, Germany, 1994.
Cardis, E., Gilbert, E.S., Carpenter, L.: Radiat. Res. 142 (1995) 117.
Cox, R., Muirhead, C.R., Stather, J.W., Edwards, A.A., Little, M.P.: Docs NRPB 6 (1) (1995).
EURATOM.: Basic safety standards for the protection of the health of workers and the general
public against the dangers arising from ionizing radiation. Council Directive 96/29/Euratom at
13 May 1996. Off. J. Eur. Commun. L159 39.
IAEA.: International basic safety standards for protection against ionising radiation and for the
safety of radioactive sources. Jointly sponsored by FAO, IAEA, ILO, NEA/OECD, PAHO and
WHO. Vienna, IAEA, Safety Series No. 115, 1996.
Pierce, D.A., Shimizu, Y., Preston, D.L., Vaeth, M., Mabuchi, K.: Radiat. Res. 146 (1996) 1.
Delongchamp, R.R., Mabuchi, K., Yoshimoto, Y.: Radiat. Res. 147 (1997) 385.
Doll, R., Wakeford, R.: Br. J. Radiol. 70 (1997) 130.
BEIR VI.: Effects of exposure to Radon. Washington DC: National Academy Press, 1988.
Mill, A.J., Frankenberg, D., Bettega, D., Hiever, L., Saran, A., Allen, L.A.: J. Radiol. Prot. 18
(2) (1998) 79.
Boice, J.D., Miller, R.W.: Teratology 59 (1999) 227.
Muirhead, C.R., Goodhill, A.A., Haylock, R.G.E.: J. Radiol. Prot. 19 (1999) 3.
UNSCEAR: Sources and effects of ionizing radiation. 2000 Report to the general assembly, with
scientific annexes. Vol II: Effects, 2000.
UNSCEAR: Hereditary effects of radiation. 2001 Report to the general assembly, with scientific
annex, 2001.
ICRP. Biological Effects after Prenatal Irradiation (Embryo and fetus). ICRP Publication 90.
Annals of the ICRP No. 1-2 Oxford, Pergamon Press (2003).

Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-1

3 Physical fundamentals

This chapter introduces the basic properties of atomic nuclei, their structure characteristics and
transformations by radioactive decay and nuclear reactions. The particle and electromagnetic radiation
associated with nuclear transformation is classified and their interaction with matter briefly described.
Special emphasis is put on fission products leading to the radioactive inventory of nuclear reactors.

3.1 Natural radioactivity


Radioactivity was observed for the first time in 1896, when Henri Becquerel [96Bec] in Paris found that
photographic plates were blackened even in the absence of light, if they were in contact with uranium
containing minerals. In 1898 the same observation was made for thorium by Marie Curie [98Cur] in
France and by G. C. Schmidt [98Sch] in Germany. Marie Curie found differences in the radioactivity of
uranium and thorium and concluded that these elements must contain unknown radioactive elements.
Together with her husband, Pierre Curie, she discovered polonium in 1898, and radium in the same year.
More historical details can be found e.g. in [95AMC].
Since radioactive radiation cannot be detected by human senses, it is necessary to use for its detection
and the identification of radioactive substances solid, liquid, or gaseous detectors, which indicate the
radiation via its interaction with the respective materials.
Radioactive elements are widely distributed in the earth's crust in more or less small concentrations.
They originate on one hand from extremely long-lived primordial radionuclides formed together with the
other stable elements and from their longer or shorter lived decay products like the important minerals of
uranium and thorium and their radioactive decay chains. Uranium and thorium are common elements in
nature. Their concentrations in the crust of the earth are about 4 and 13 mg/kg, respectively, and the
concentration of uranium in seawater is about 3 g/l. The most important uranium mineral is pitchblende
(U308). The most important thorium mineral is monazite, which contains between about 0.1 and 15 % Th.
Natural radioactivity is mainly observed with heavier primordial elements and seldom with light ones
(e.g. 40K and 87Rb).
On the other hand, radioactive elements can be formed - especially in case of light shorter lived
elements - by interactions of cosmic radiation with the atmosphere. This radiation entering the
atmosphere from outside originates from the sun as well as from material in the deep interstellar space. It
produces a variety of elementary particles (protons, neutrons, electrons, positrons, mesons, photons) and
of radioactive atoms via cascades of interactions with the gas molecules in the atmosphere.
Among others, 14C, 10Be , 7Be, and 3H (tritium) are produced in the atmosphere by cosmic radiation.
Examples for the production rate are: about 2.2 104 atoms of 14C per second per m2 of the earth's surface
and about 2.5 103 atoms of tritium per second and m2. Taking into account the radioactive decay and the
dwell times in the atmosphere, these data result in a global equilibrium inventory of about 6.3 104 kg of
14
C and of about 3.5 kg of 3H.
The measurement of local concentrations of natural radionuclides like 14C, 40K, 87Rb is very useful for
the determination of the age of the respective material. However, other sources have to be taken into
account like the production of these nuclides in nuclear power plants and by explosions of nuclear bombs.
Landolt-Brnstein
New Series VIII/4

3-2

3 Physical fundamentals

[Ref. p. 3-39

3.2 Elements, isotopes and radionuclides


3.2.1 Atoms, electrons and the Periodic Table of Elements
3.2.1.1 The atom
The atom is the smallest particle of an element, which can no more be subdivided by chemical methods. It
consists of a (heavy) nucleus surrounded by a number of (light) electrons. The elements differ from each
other by the structure of their atoms i.e. the composition of the nucleus and the number and distribution of
the electrons. The diameter of an atom is approximately 108 cm. The nucleus contains positively charged
protons and electrically neutral neutrons and has, therefore, a positive charge. Its diameter is in the order
of 1013 cm, which is five orders of magnitude smaller than the atomic diameter. Since atoms in their
normal state are electrically neutral, the number of (negative) electrons in the shell must balance the
number of (positive) protons in the atomic nucleus.
3.2.1.2 The electron shells
The electron distribution around the atomic nucleus is described and denoted according to Bohr's shell
model. Electrons move in discrete shells with energy states (binding energies) decreasing with increasing
distance from the nucleus. They are numerated beginning with n = 1 and denominated by capital letters K,
L, M, .... The maximum number of electrons in one shell is 2n.
By energy transfer electrons can be excited into higher lying weaker bound states. The electrons are
released into an unbound state by an energy transfer of at least the binding energy of the respective shell
leaving the residual atom in an ionised state. The remaining hole in the electron shell is filled by one of
the outer electrons releasing the energy difference in form of characteristic photon radiation
(fluorescence) or by radiationless energy transfer to another bound electron, which in turn is emitted
(Auger electron).
3.2.1.3 The Periodic Table of Elements
When ordering all elements according to their atomic weight, one finds a periodical repetition of special
properties leading to several groups of elements with a remarkable similarity of their chemical and
physical properties. Within these groups very similar differences of the atomic weights of neighbouring
group members are found. The most outstanding element group is formed by the noble gases, similar
observations are made for the halogens fluorine, chlorine, bromine, and iodine, as well as for the
chalcogens sulphur, selenium, and tellurium. First indications of this periodical behaviour were already
described in the early 19th century. In 1869 L. Meyer and D. Mendeleev succeeded in placing all known
elements satisfactory in the periodical system.
In this periodical system all elements, except of the rare earth elements (lanthanides) and the
transuranium elements (actinides) are sorted into 6 periods. This ordering shows two short periods with 8
elements each and three long periods comprising 18 elements, taking into account some not naturally
occurring elements, like technetium. The three long periods are subdivided into two parts. Between these
one finds three triples of very similar elements: Fe-Co-Ni, Ru-Rh-Pd, Os-Ir-Pt, respectively. In this
arrangement chemically similar elements form columns combined in 8 groups, subdivided into main
groups and auxiliary groups of transition elements. The 8th main group containing the noble gases by
convention is called group 0.
This ordering was explained later on by Bohr's atomic model. The periods correspond to the filling of
the different electronic shells, which can take up 2n electrons each. The noble gases in group 0
correspond to a completely filled shell in each period.
Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-3

3.2.2 Atomic nuclei, nuclides and the Chart of Nuclides


3.2.2.1 Nuclei and nuclides
Atomic nuclei are composed of protons and neutrons having the common denotation nucleons. The
corresponding number of protons Z is called the atomic number. It characterizes the element and its
chemical behaviour, while the physical properties are defined by Z as well as by the neutron number N.
Atoms with a defined nucleus and the corresponding number of electrons are called nuclides. Each
nuclide is identified by its mass number A = Z + N and is marked by the following denotation for the
element symbol El:
A
A
4
235
He = He - 4 = 2 He42 , 235U = U - 235 = 92 U143
Z El N or in shorter form El (or El-A), e.g.:
since Z defines uniquely the element and N is given as well by N = A Z.
The following denotation is used for neighbouring nuclides (cf. Fig. 3.1):
- nuclides with equal

atomic number Z are called isotopes


mass number A are called isobars
neutron number N are called isotones
- nuclides in long-lived excited states are called isomers.
Different isotopes of an element exhibit almost the same chemical behaviour (very small differences
are observable in special experiments only: isotopic effect). In general, the elements represent a mixture
of all of their stable isotopes.

Atomic number Z

Isobars

Isotones

49
48
47

74
Isotopes

45
44

72

43
60

62 64 66 68

70

Fig. 3.1. Representation of nuclides in a chart of nuclides as


proposed by E. Segr and definition of isotopes, isotones, and
isobars.

Neutron number N

Today nearly 3000 nuclides are known most of which are extremely short-lived and can be produced
in complicated experiments only. More than 300 nuclides are found in nature and can be assigned to one
of four categories:
1. 258 stable nuclides
2. 25 nuclides with Z <80 having extremely long half-lives (radioactive decay not proved uniquely in all
cases)
3. about 15 quasistable nuclides with half-lives >105 years (including 238U, 235U, 232Th and 244Pu)
existing since the genesis of the elements and called primordial radionuclides. Among these 238U,
235
U and 232Th and their radioactive decay products form the main sources of natural radioactivity
4. radionuclides continuously produced by the impact of cosmic radiation: e.g. 14C, 10Be, 7Be, 3H.
3.2.2.2 The Chart of Nuclides
In order to get a synopsis of this vast amount of nuclides several types of schematic representations have
been introduced in the past. The most widespread scheme has been proposed by E. Segr and has been
realized among others in the Karlsruhe Chart of Nuclides [98PKS]. In this scheme the nuclides are
arranged in such a way (cf. Fig. 3.1) that the proton number Z is given on the ordinate and the neutron
number N = A Z on the abscissa, respectively. Each experimentally observed nuclide is represented by a
square containing the symbol of the element and the number of nucleons (mass number) and in addition
Landolt-Brnstein
New Series VIII/4

3-4

3 Physical fundamentals

[Ref. p. 3-39

various decay or structure data. For example, in the Karlsruhe Chart of Nuclides [98PKS] besides the
half-lives, the decay modes, the types of emitted radiation and the energies of the most abundant radiation
types are given as observable in respective experiments. Other charts, nowadays often distributed in
electronic form (e.g. [99Mag]), are based on (evaluated) nuclear data files enabling access to all known
decay data of all known radionuclides.
In these charts the stable nuclides lie around a central branch with approximately N = Z for the lighter
nuclides. With increasing mass this branch flattens due to the increasing neutron excess necessary for the
stability of heavier nuclei. This branch (valley of beta-stability) separates the neutron rich nuclides
undergoing -decay from the proton rich nuclides undergoing +-decay. Furthermore, with increasing
distance from the valley of stability, i.e. with increasing neutron- or proton excess, the stability and
consequently the half-lives of the nuclides decrease drastically. Thus, radionuclides with half-lives in the
range of hours or longer are only found in narrow bands along the valley of stability.
The so called strong interaction, by which the nucleons are bound in the atomic nucleus, is a very
short ranged pairing force binding the nucleons very close together. This leads to a very high density of
the nuclear matter. With increasing mass number stable nuclei exhibit an increasing neutron excess (up to
50 % for the heaviest nuclei) necessary to balance the increasing repulsive force of the positively charged
protons.
Nuclei like the electron shells can be excited by energy transfer into higher energetic states. The
de-excitation of such states occurs under emission of characteristic nuclear radiation, which is in general
-radiation. At sufficiently high excitation energies (above the separation energy of nucleons) the
emission of nucleons or nucleon clusters becomes possible.

3.3 The structure of the atomic nucleus


3.3.1 Elementary particles
Though the modern high energy and particle physics deals with elementary particles far below the level
of those forming the atomic nuclei, only the latter shall be discussed here and are according to classical
nuclear physics denominated as elementary particles of the nucleus, the nucleons.
3.3.1.1 Charge, mass and stability of the nucleons
The proton is stable and has a positive electric charge equal and opposite to the elementary charge. The
neutron is electrically neutral and its mass is slightly larger than that of the proton. The free unbound
neutron is unstable and decays with a half-life of 10.25 min into a proton and an electron and an antineutrino.
The following units are used for atomic and nuclear quantities (see also Table 3.1), making use of the
energy-mass equivalence in Einstein's equation E = mc2, which leads to the correspondence 1 kg =
5.6095892 1029 MeV:
elementary charge:
e = 1.6021764621019 C
mass:
atomic mass unit: u = 1.660538731027 kg = 9.31494013102 MeV/c2, where the basis of
this unit is 1/12 of the mass of the neutral 12C atom (including the
masses of its 6 electrons).

Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-5

Table 3.1. Frequently used general constants (main values taken from [99MoT]) and conversion factors.
The standard deviations are given in parentheses.
Symbol
c
h
= h/2
u
mn

value

quantity
1

299792458 m s
6.62606876 (52)1034 J s
1.054571596 (82)1034 J s
1.66053873 (13)1027 kg
9.31494013 (37)102 MeV/c2
1.00866491578 (55) u

speed of light
Planck constant
atomic mass unit
neutron rest mass

27

mp

1.67492716 (13)10 kg
9.39565330 (38)102 MeV/c2
1.00727646688 (13) u

proton rest mass

27

me
e
e/me
re = e2/mec2
a0

R
NA
Vm
R
k = R/NA
F = NAe

1.67262158 (13)10 kg
9.38271998 (38)102 MeV/c2
9.10938188 (72)1031 kg
0.510998902 (21) MeV/c2
1.602176462 (63)1019 C
4.803204197 (19)1010 esu
1.758820174 (71)1011 C kg1
5.27281023 (21)1017 esu g1
2.817940285 (31)1015 m
5.291772083 (19)1011 m
7.297352533 (27)103
1.0973731568549 (83)107 m1
6.02214199 (47)1023 mol1
2.2413996 (39)102 m3 mol1
8.314472 (15) J mol1 K1
8.314472 (15)107 erg mol1 K1
1.3806503 (24)1023 J K1
8.617342 (15)105 eV K1
9.64853415 (39)104 C mol1
2.892557769 (12)1014 esu mol1

electron rest mass


elementary charge
specific electron charge
electron radius
Bohr radius
fine structure constant
Rydberg constant
Avogadro constant
molar volume of an ideal gas at s. t. p.
molar gas constant
Boltzmann constant
Faraday constant

Some useful conversions:


1 Ci = 3.71010 Bq (= disintegrations s1)
1 W ^ 3.11010 fissions s1
1 MWd ^ 2.71021 fissions ^ 1g fissionable material

Landolt-Brnstein
New Series VIII/4

1 A = 6.2415097451012 e s1
1 eV/Atom ^ 23 kcal mol1
1 MeV Ci ^ 5.93103 W

3-6

3 Physical fundamentals

[Ref. p. 3-39

Average binding energy per nucleon B / A [MeV ]

9
8
7
6
5
4
3
2
0

Fig. 3.2. Average binding energy per nucleon in dependence on the mass number.
8 16 24 30

60 90 120 150 180 210 240


Mass number A

3.3.1.2 Binding energy


The experimentally measured atomic weights M of stable pure isotopes are not integer multiples A of the
atomic mass unit u. This deviation is called the mass defect M = M Au. In addition, also the mass
M(Z, A) of a nucleus differs from the mass sum of its nucleons and is always smaller: M(Z, A) < ZMp +
NMn.
According to Einstein's equation, the mass difference M = ZMp + NMn M(Z, A) corresponds to an
energy B = Mc2 = [ZMp + NMn M(Z, A)]c2, which is necessary to separate all nucleons of the
nucleus. B is called the total binding energy of the nucleus and is always positive for stable nuclides.
B/A is the average binding energy per nucleon, which has a mean value of about 8 MeV/nucleon,
except for the very light and heavier elements (cf. Fig. 3.2). This behaviour has the most important
consequence that energy can be gained by fission of heavy as well as by fusion of light nuclei, both
leading to a higher energy for the product(s).

3.3.2 Nuclear transformations


3.3.2.1 Nuclear reactions
The vast amount of radioactive nuclides as shown in charts of nuclides do not occur in nature but can only
be produced artificially by means of nuclear reactions. In a nuclear reaction a normally stable nuclide is
bombarded with a beam of one of many types of radiation (charged particle or neutron radiation,
electromagnetic radiation, electrons).
The simplest form of a nuclear transformation is the radioactive decay caused by internal excess
energy: A B + b + E. This is a mononuclear reaction. Binuclear reactions, denoted generally as
nuclear reactions, are induced by bombarding target nuclei with a beam of specific projectiles. They are
described by: A + a B + b + E, where A is the target nuclide, a the projectile, B the product nuclide,
and b the ejectile (particle or photon emitted). The energy E is also called the Q value of the reaction.
The first nuclear reaction was observed in a cloud chamber in 1919 by Rutherford:
14

N + 4He 17O + 1H, in shorter form


N(,p)17O or generally
A(a,b)B.
14

Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-7

When comparing chemical and nuclear reactions one finds the following main differences:
In chemical reactions macroscopic amounts of matter as a whole are altered, in nuclear reactions
individual atoms.
In chemical reactions the total involved mass is conserved, in nuclear reactions the sum (E + mc2) of
the energies and the mass-energy equivalents.
The energies of chemical reactions are relatively small and comparable with the energies of chemical
bonds (of the order of eV), whereas the energies involved in nuclear reactions are about 6 orders of
magnitude higher (of the order of MeV).
Many nuclear reactions pass over a transition state, similarly to chemical reactions:
A + a (C)* B + b + E
The transition state (C)*, which is excited to an elevated energy, is also called a compound nucleus; its
lifetime is very short (<1013 s).
While the probability for a mononuclear reaction (i.e. for radioactive transmutation) is given by the
decay constant , in the compound nucleus model the reaction probability is determined by both the
probability that the projectile a will react with the nuclide A and by the probability that the nuclide B is
produced.
The time duration of a nuclear reaction is in between about 1023 and 1013 s. The lower limit is given
by the crossing time of a particle passing the nucleus with the velocity of light, and the upper limit holds
for slow reactions, e.g. with thermal neutrons.
3.3.2.2 Projectiles for nuclear reactions
Charged particles
Positively charged particles inducing nuclear reactions, such as protons, deuterons or ions with higher
atomic numbers Z, need a minimum energy to surmount the repulsive Coulomb barrier formed by the
protons of the nuclei. The Coulomb barrier U can be calculated approximately from the equation
U

Z A Za e2
Z Z
1/ 3 A a 1/ 3
4 0 r
AA + Aa

(3.3.1)

where ZA and Za are the charge numbers of the nuclide A and the projectile a, respectively, e is the unit
charge, 0 the electric field constant, and r the distance, which in this approximation is set to be
r r0(AA1/3 + Aa1/3)

(3.3.2)

AA and Aa are the mass numbers of the nuclide A and the projectile a, respectively. For the Coulomb
barrier U the following approximate values can be calculated:

U 1.8 MeV for the reaction of a proton with 12C,


U 13 MeV for the reaction of a proton with 238U,
U 24 MeV for the reaction of an -particle with 238U,
U 130 MeV for the reaction of 12C with 238U and
U 700 MeV for the reaction of 238U with 238U.

With increasing atomic numbers the approximate formula (3.3.1) becomes incorrect. For the reaction
of 238U with 238U, e.g., a more exact value for the Coulomb barrier is U 1500 MeV.

Landolt-Brnstein
New Series VIII/4

3-8

3 Physical fundamentals

[Ref. p. 3-39

Charged particles for nuclear reactions originate from particle accelerators. They are produced in ion
sources by bombarding a gas with energetic electrons. The positive ions are extracted by means of an
electrode. For the further acceleration various set-ups are used. The two main groups are linear
accelerators (linacs) with single (e.g. Cockroft-Walton type) and multiple (e.g. tandem Van de Graaff
accelerators) stages and circular accelerators. With single-stage linear accelerators proton or deuteron
beams of up to about 10 mA with energies of up to about 4 MeV are obtained. These accelerators are
often applied as injectors in larger machines for the production of high-energy particles. In two-stage
tandem Van de Graaff accelerators proton beams of about 20 MeV and -particles of about 30 MeV are
obtained. In addition, lighter heavy ions can be efficiently accelerated, too. The intensity of the beam
current varies between about 10 and 100 A.
In cyclotrons the ions move on spiral paths with increasing radius guided by a suitable magnetic field,
while in synchrotrons the circular orbit of the particles remains constant and the guiding magnetic field
increases with the momentum of the ions.
In modern machines, protons, deuterons and -particles can be accelerated to energies of several 100
MeV up to about 1 GeV. For the production of radionuclides, relatively small accelerators are used
producing particle energies of 10 to 30 MeV and ion currents of the order of 100 A.
With light charged particles mainly nuclides on the left hand side of the valley of -stability are
produced (+- and electron capture activities). Increasing projectile energies lead then to an increasing
neutron deficit of the produced radionuclides.
Heavier ions are produced in special types of linear or circular accelerators. The term heavy ions is
used in this context for all ions heavier than -particles and includes light ions e.g. of lithium, carbon or
oxygen as well as heavy ions of elements up to uranium. These projectiles are widely used in basic as
well as in applied research and techniques. Due to the great number of nucleons, which they can transfer
to the target nuclide, the main application fields of heavy ions are: synthesis of new (including the socalled super-heavy) elements, production of nuclides far off the line of -stability, investigation of
nuclear matter at high densities, production of small holes with defined diameters in thin foils and
irradiation of tumors in medicine.
Neutrons
Neutrons are frequently used projectiles for nuclear reactions. Since they do not carry a positive charge,
they do not experience Coulomb repulsion, and even low-energy (thermal and slow) neutrons can easily
enter the target nuclei. Neutrons with energies of the order of 1 to 10 eV (resonance neutrons) exhibit
relatively high absorption maxima. Furthermore, neutrons are available in large quantities in nuclear
reactors with fluxes of the order of about 1010 to 1016 cm2 s1.
Neutrons may also be produced by nuclear reactions, such as 9Be(,n)12C, 9Be(d,n)10B, 9Be(,n)2,
d(,n)p, d(d,n)3He or t(d,n). Alpha particles are available from radionuclides like 226Ra or 210Po, and RaBe neutron sources have formerly often been applied in experimental neutron physics. Gamma-rays of
sufficiently high energy for (,n) reactions may be supplied by radioactive nuclides, such as 124Sb.
Neutrons are also available from spontaneously fissioning nuclides like 252Cf (1 g of 252Cf emits
2.3 106 neutrons per second). In neutron generators deuterons with energies between 0.1 and 10 MeV
are produced in small accelerators and impinge on a suitable target, e.g. a tritium target. The neutron
fluxes available from neutron sources and neutron generators vary between about 105-108 and 108-1011
cm2s1, respectively. Neutrons produced by the reaction t(d,n) have energies of about 14 MeV.
Reactions with neutrons are used to produce nuclides on the right-hand side of the line of -stability,
i.e. nuclides undergoing -decay.

Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-9

Electrons and -rays


For the acceleration of electrons to high velocities approaching the velocity of light, much less energy is
needed than for the acceleration of ions. They are accelerated in linear or circular (betatrons,
synchrotrons) accelerators. Depending on the size of the installations, energies of the order of 10 to 300
MeV are obtained in linear accelerators and betatrons, whereas in electron synchrotrons electrons with
energies up to the order of 10 GeV are available.
Electrons are not directly used for the production of radionuclides. However, the high-energetic
bremsstrahlung, which is emitted, when the electrons are slowed down in a target of high atomic number,
is applied for the induction of nuclear reactions (photo-nuclear reactions). The maximum energy of this
bremsstrahlung corresponds to the energy of the incident electrons and is in the range of high-energy
-rays. Gamma rays are also available from -emitters, such as 60Co or 124Sb, but with comparably lower
energies and intensities.
The monoenergetic synchrotron radiation emitted by electrons moving in the magnetic field of a
synchrotron has found many applications because of the high intensities, broad energy ranges, and small
beam diameters available. The energy of the synchrotron radiation is in the range of the energy of X-rays.
3.3.2.3 Artificial radionuclides for medical and technical applications
Numerous radionuclides are especially produced at accelerators or nuclear reactors for dedicated
applications in medicine and technology. In particular, in nuclear medicine new diagnostic methods have
been established and are applied with increasing frequency such as single photon emission computed
tomography (SPECT) and positron emission tomography (PET). The main advantage of these methods as
compared to other imaging processes is their capability not only to provide an image of inner organs but
also to give information about their function (metabolism). These methods use short living accelerator
isotopes like 11C, 13N, 18F, 123I or 153Gd. The diagnostical goals are detection of malfunction or cancer
stroke of thyroid gland, kidneys, liver, heart and brain.
For so-called targeted radiotherapy of cancer various -emitters e.g. 90Y, 125I and 107Pd are being used.
Quite recently, -emitters like 211At, and 213Bi/225Ac are under development for such purposes which
promise to have thousands fold better biological effectiveness.
Technical application of primordial (241Am) and artificial radionuclides (e.g. 55Fe, 57, 60Co, 137Cs) range
from smoke detection, measurement of moisture content in materials via thickness measurements for
various materials to wear diagnostics of machine parts (57Co). The latter method is industrially applied by
many car factories.
3.3.2.4 Excited states, level- and decay schemes
Like the electrons, which can occupy only discrete states in the shell with defined energies, also the
nucleons in the atomic nucleus are in discrete states with well-defined energies. The state with the lowest
energy is called the groundstate, its energy is per definition set to zero and all higher lying states (excited
levels) are referred to this groundstate.
In many cases of radioactive decay and even more often after nuclear reactions the product of the
transformation is not in the groundstate but rather in an excited state. These and other excited states
correspond to a series of defined energy levels, which are specific for each nuclide. They release their
excitation energy by emission of one or several -rays, mostly within about 1013 s after their formation
by a nuclear reaction or by a preceding - or -decay. In some special cases, e.g. for very low transition
energies, -transitions can be hindered (forbidden transitions) resulting in longer half-lives of these
metastable isomeric states.
In order to visualize the level structure of a nuclide, the levels are ordered in a scheme according to
their energy above the ground state and are denoted besides the energy by the specific quantum
numbers nuclear spin and parity, resulting from the nucleons contributing to the excitation. In addition,
Landolt-Brnstein
New Series VIII/4

3-10

3 Physical fundamentals

[Ref. p. 3-39

the assigned -transitions and for isomeric states the half-lives are indicated. In the decay schemes the
nuclides with higher atomic number are ordered to the right hand side and the decay types and transition
energies to the levels in the daughter nuclide are specified.
The construction of level schemes is performed either directly by sophisticated nuclear reaction
experiments and/or by a comprehensive investigation of the radioactive decay of the mother nuclide
feeding several states with subsequent -decay. While for light radionuclides the decay schemes are
mostly relatively simple due to the fact that only few levels can be fed by the decay of the mother nuclide,
heavier nuclides exhibit mostly very complicated schemes with many levels and -transitions.

3.4 Radioactive decay


3.4.1 Basic properties
3.4.1.1 Decay law for a single radionuclide
From the observation that the number of nuclei decaying per unit time is proportional to the actual
number N of radioactive nuclei the following time law results:
dN/dt = N

(3.4.1)

The constant of proportionality is characteristic for each nuclide and is called decay constant. It
corresponds to the fraction of the actually present nuclei, which decays per unit time.
The integration of equation (3.4.1) results in the general decay law:
N(t) = N(0)et

(3.4.2)

The number of atoms of a nuclide decaying per unit time is called its decay rate (N). It is measured
in Bequerel (1Bq = 1/sec). The formerly used unit of radioactivity was the Curie (1 Ci corresponding to
3.7 1010 Bq). A specified activity refers to the respective nuclide only, independent of possible
subsequent radioactive decays of daughter nuclides.
The decay rate N is usually set equal to the activity A. In general, however, an experimentally
measured activity A is lower but always proportional to the decay rate of a sample A = FN. This is due
to the fact that the measured rate depends on the experimental setup, the sensitivity of the detector used
and the thickness, backing and area of the sample. In the following, this factor F will be set to 1.
The general decay equation becomes therefore:

N(t) = N(0)et

(3.4.3)

A(t) = A(0)et

(3.4.4)

or
and
ln A(t) = ln A(0) t

(3.4.4a)

where A(t) means the activity at time t and A(0) the activity at time t = 0.
If the original number of atoms N(0) has decreased to one half, N(t) = N(0)/2 and t = T (= T: halflife), equ. (3.4.2) gives = ln 2/T 0.693/T. With this expression for it follows from equ. (3.4.4a):
ln A(t) = ln A(0) ln(2)t/T

(3.4.4b)
Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-11

The use of the half-life is often more convenient than that of the decay constant . Hence, in final
expressions is replaced by ln 2/T. Half-lives are usually given in units of seconds (s), minutes (m),
hours (h), days (d) and years (y).
Examples of radioactive decays into a stable final state are: 60Co, 128I, ...
3.4.1.2 Branching radioactive decay
Several nuclides can decay via more than one decay branch, e.g. by electron capture as well as by
or +-decay. The decay constant being characteristic for the nuclide's decay is in this case composed
of the partial decay constants of the individual decay branches:

= 1 + 2 + ... + n

(3.4.5)

Since the activity for each decay branch is Ai = iN, the total activity of the nuclide is given by:
or

A = A1+ A2 + ... + An = 1N + 2N + ... + nN

(3.4.6)

A(t) = 1N(0) et + 2N(0) et + ...+ nN(0) et

(3.4.7)

The partial activities are proportional to the total number N of the respective nuclide decreasing with
its corresponding half-life.
Example: 64Cu 64Ni + 64Zn (T=12.7 h; +=19 %, =39 %, EC = 42 %)
3.4.1.3 Mixture of several nuclides without genetic relations
The total activity of a mixture of radionuclides at any time is proportional to the sum of the individual
activities at the respective time:
A = A1 + A2 + ...+ An = A1(0)e1t + A2(0)e 2t + ...+ An(0)e nt

(3.4.8)

3.4.1.4 Activity of nuclides with genetic relations


For activity chains with generic mother-daughter relations the activity of the mother nuclide (index 1) is
given by equ. (3.4.4). For the determination of the daughter activity (index 2) one has to consider both the
formation via decay of the mother nuclide with the decay constant 1 and the decay of the daughter with
its own decay constant 2:
dN2/dt = 1N1 2N2

(3.4.9)

The number of daughter nuclei at time t is:


N 2 (t ) = N1 (0)

(e

1t

2t

) + N ( 0) e

2t

2
2 1
2
t
t
t
A2 (t ) = A1 (0)
e 1 e 2 + A2 (0)e 2 or
2 1

A2 (t ) = A1 (0)

T1
T1 T2

t ln 2
t ln 2
t ln 2

e T1 e T2 + A (0) e T2
2

If at t = 0 the daughter activity is zero, equ. (3.4.10b) becomes


Landolt-Brnstein
New Series VIII/4

(3.4.10)
(3.4.10a)

(3.4.10b)

3-12

3 Physical fundamentals

T1
e
A2 (t ) = A1 (0)
T1 T2

t ln 2
T1

t ln 2
T2

[Ref. p. 3-39

(3.4.11)

3.4.1.5 Secular radioactive equilibrium (T1 T2)


If the half-life T1 of the mother nuclide is much larger than that of the daughter nuclide T2 the latter can be
discarded in the quotient of equ. (3.4.11):
t ln 2
t ln 2

T1

e T2
A2 (t ) = A1 (0) e

(3.4.12)

If in addition t T1, equ. (3.4.12) becomes:


t ln 2

A2 (t ) = A1 (0) 1 e T2

(3.4.13)

what means that the increase of the daughter activity depends only on T2.
After a period of about 6 - 7 T2 the daughter activity equals approximately that of the mother nuclide.
The total activity for periods small when compared to T1 is constant and equals 2A1.
Example: 90Sr 90Y 90Zr (T1=28.64 y, T2=64.1 h)
3.4.1.6 Transient equilibrium (T1 > T2)
If T1 > T2 but both are of the same order of magnitude equ. (3.4.11) is valid. It can be simplified for
periods t larger than about 6-7 T2, since the expression

t ln 2
T2

decreases much faster with time than

t ln 2
T1

T
A2 (t ) = A1 (0) 1 e
T1 T2

t ln 2
T1

(3.4.14)

The system reaches an equilibrium state, where the daughter activity is


A2 =

T1
A1
T1 T2

and decreases with the half-life of the mother T1.


The daughter activity A2 for t = 0 equals zero and approaches zero again for very large times t. The
time tm, where A2 has a maximum, is found from the first derivative of equ. (3.4.11):
Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

tm =

3 Physical fundamentals

T1T2
T
ln 2
(T2 T1 )ln 2 T1

3-13
(3.4.15)

For the total mother- and daughter activity

Atotal = A1 + A2 = A1 (0)e

t ln 2
T1

+ A1 (0)

T1
T1 T2

t ln 2

e T1 e

t ln 2
T2

(3.4.16)

the time for the maximum is found correspondingly:


tm =

T1T2
T2
ln
(T2 T1 ) ln 2 2T1T2 T2 2

(3.4.17)

Example: 132Te 132I 132Xe (T1=76.3 h, T2=2.3 h)


3.4.1.7 Similar half-lives (T1 T2)
If the half-lives of mother and daughter nuclide are very similar, equ. (3.4.11) can no more be used. By
means of series development one obtains:
A2 (t ) = A1 (0)

t ln 2
e
T2

t ln 2
T2

1 +

t (T1 T2 )ln 2

2T1T2

(3.4.18)

The term
t (T1 T2 )ln 2
2T1T2

can be neglected if it adopts values 1. Equation (3.4.18) has then the following form:
A2 (t ) = A1 (0)

tln 2
e
T2

t ln 2
T2

(3.4.19)

For
t (T1 T2 )ln 2
1
2T1T2

the bracket in equ. (3.4.18) cannot be neglected. For


t (T1 T2 )ln 2
>1
2T1T2

instead of equ. (3.4.18) equ. (3.4.11) has to be used again. For the special case T1 = T2 one obtains
formally again equation (3.4.19).

Landolt-Brnstein
New Series VIII/4

3-14

3 Physical fundamentals

[Ref. p. 3-39

From the first derivative of equ. (3.4.19) one gets the time tm of the maximum activity:
tm =

T
1
=
ln 2

(3.4.20)

If the mother nuclide at time t = 0 did not contain any daughter activity, tm corresponds to the time
where mother and daughter activities are equal.
Example: 101Mo 101Tc 101Ru (T1=14.6 m, T2=14.2 m)
3.4.1.8 Half-life of mother nuclide shorter than half-life of daughter (T1 < T2)
In case of T1 < T2 equ. (3.4.11) can be rewritten as:

T1
A2 (t ) = A1 (0)
e
T2 T1

t ln 2
T2

t ln 2
T1

(3.4.21)

In this equation T1/(T2T1) <1, if 2T1 < T2. For times t T2 the increase of the daughter activity A2
depends only on the half-life T1 of the mother nuclide in contrast to the cases of secular and transient
equilibrium:

A2 (t ) = A1 (0)

T1
1 e
T2 T1

t ln 2
T1

(3.4.22)

For times t > T1 the daughter activity decreases with its individual half-life:

T1
A2 (t ) = A1 (0)
e
T2 T1

t ln 2
T2

(3.4.23)

In this case no equilibrium is reached, since the ratio of daughter and mother activities increases
permanently as function of time:
A2 (t )
T1 T1T2
e
=
A2 (t ) T2 T1

T2 T1

t ln 2

(3.4.24)

The daughter activity passes a maximum, like in the cases 3.4.1.6 and 3.4.1.7. The corresponding time
tm can be calculated from equ. (3.4.15), too.
Example: 135I 135Xe 135Cs (T1=6.61 h, T2=9.1 h)
3.4.1.8 Successive activities in longer decay chains (radioactive families)

In case of further decay of a daughter nuclide (decay chains, radioactive families) the activity A3 at time t
of the next member in the chain is given by equ. (3.4.25) provided that at time t = 0 only the mother
activity existed:

Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

t ln 2
t ln 2

T3
T2
T1
e
e T2 +
e T1 +
A3 (t ) = A1 (0)T1
(T1 T2 )(T1 T3 )
(T2 T1 )(T2 T3 )
(T3 T1 )(T3 T2 )

3-15
t ln 2
T3

(3.4.25)

Example: 211Pb 211Bi 207Tl 207Pb (T1=36.1 m, T2=2.17 m; T3=4.77 m)


For T1 T2 and T1 T3 and for times t T1 equ. (3.4.25) is simplified to:

T2
e
A3 (t ) = A1 (0)1
T2 T3

t ln 2
T2

T3
e
T2 T3

t ln 2
T3

(3.4.26)

For T2 > T1 and T2 > T3 and for times t T1 and t T3 equ. (3.4.25) is further simplified to:

T1T2
e
A3 (t ) = A1 (0)
(T2 T1 )(T2 T3 )

tln 2
T2

(3.4.27)

and for T1 < T2 and T3 T2 to:


A3 (t ) = A1 (0)

T1
e
T2 T1

t ln 2
T2

(3.4.28)

= A2 (t )

The general equation for the activity of the nth member of a radioactive decay chain was given by
Bateman [10Bat] for the case that at t = 0 there are no decay products of the mother nuclide:

A n (t ) = A1 (0)T1 C1e

t ln 2
T1

+ C2e

t ln 2
T2

+ ...... + C n e

t ln 2
Tn

(3.4.29)

with
Cn =

(n 2 )

(n 2 )

(n 2 )
Tn
T1
T2
, C2 =
, C1 =
(Tn T1 )(Tn T2 )...(Tn Tn 1 )
(T2 T1 )(T2 T3 )...(T2 Tn )
(T1 T2 )(T1 T3 )...(T1 Tn )

3.4.2 Decay modes


In order to minimise their total energy, many nuclides undergo radioactive decay. Depending on the
lowest reachable energy state various decay types occur either caused by the strong interacting nuclear
force (non-isobaric decay with change of the mass number) or by the weak interaction (isobaric decay
with constant mass number).
The most common radioactive decay modes observed for naturally occurring nuclides are -decay
(non-isobaric), -decay (isobaric) in the 3 types -, +-decay, and electron capture, and -decay
(electromagnetic transitions between nuclear states). Much less abundant decay modes are the
spontaneous fission and the emission of nucleons (protons, neutrons) and nucleon clusters. The following
scheme (Fig. 3.3) shows the nuclear transmutations (changes of atomic and mass number) originating
from the main decay modes. A more complete compilation is given in Table 3.3.

Landolt-Brnstein
New Series VIII/4

3-16

3 Physical fundamentals

Z, A

Z2 , A 4

Z1 , A

Z, A

Z, A

Z, A

EC

Z+1 , A

Z, A

[Ref. p. 3-39

Fig. 3.3.
Transmutations of
atomic and mass numbers during
the main nuclear decay modes.

During the -decay 4He2+ ions are emitted by the unstable radionuclide, whose mass and atomic
numbers decrease consequently by 4 and 2 units, respectively. The transitions take place between well
defined states in the mother and daughter nuclides leading to one or several monoenergetic -lines for
each -decaying nuclide. The energy available for the -decay can be obtained from the Einstein relation
and the average binding energy per nucleon (Fig. 3.2) resulting in the fact that all nuclides with A > 140
should be unstable against -decay. However, further effects have to be considered. In order to leave the
nucleus, the -particle has to surmount a high potential barrier, which can be passed at high energies
only. Nevertheless, -particles are observed with energies well below this barrier. This is a consequence
of the quantum mechanical tunnel effect according to which a certain probability exists to pass this barrier
at lower energies. Since the width of this barrier decreases with increasing potential, one could expect that
-particles with higher energy tunnelling at a narrower barrier width would pass the barrier with higher
probability. This was observed phenomenologically already in 1911 by Geiger and Nutall who found a
clear correlation between half-life and range of -particles, the latter being a direct measure for the energy: log = alog E + b (Geiger-Nutall rule).
Table 3.2. Compilation of nuclear decay modes
Decay mode
Symbol Radiation emitted
helium nuclei He2+
-decay

Decay process
(Z, A)(Z2, A4) + 4He2+

electrons e, antineutrinos

(Z, A)(Z+1, A) + e +
e

positrons e+, neutrinos

(Z, A)(Z1, A) + e+ + e

electron capture (EC)

-transition

isomeric transition (IT)

internal conversion (IC)

proton decay
spontaneous fission

p
sf

characteristic X-rays/ Auger (Z, A)(Z1, A) + e


electrons of the daughter
nuclide, neutrinos
release of nuclear excitation
photons (h)
energy after particle emission
delayed release of nuclear
photons (h)
excitation energy
conversion electrons and
transfer of nuclear excitation energy
characteristic X-rays
to an inner shell (K-,L-,...) electron
protons
(Z, A) (Z1, A1) + p
fission products, neutrons
(Z, A) (Z', A') + (ZZ', AA'x)
+xn

-decay

The denotation -decay combines all nuclear decay modes, by which the atomic number Z is changed
by one unit, while the mass number A remains unchanged (cf. Fig. 3.3). During -decay a neutron in the
nucleus is transformed into a proton, a negative electron together with an (anti-)neutrino is emitted and
the atomic number changes from Z to Z+1. In contrast, during +-decay a proton in the nucleus is
transformed into a neutron, a positive positron together with a neutrino is emitted and the atomic number
changes from Z to Z1. From the mass balance of this process it follows that it can take place only, if the
atomic masses of mother and daughter nuclide differ by at least two electron rest mass units. The
neutrinos emitted together with the -particles are uncharged nearly mass-less particles which give rise to
the continuous form of the -spectra, since the available transition energy is shared between the
-particle and the usually not observed neutrino. The third possibility competing to the +-decay, the
electron capture, results from the fact that the electron orbits of the inner shells - especially the K-shell Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-17

can pass the nucleus and the electron can be captured by an excess proton forming a neutron. The hole in
the electron shell is filled by an electron from outer shells giving rise to the emission of characteristic
X-rays of the daughter nuclide, which are the only external radiation of this process.
5+
60

5.272y

Co
E = 315 keV
4+

2505
E = 1173.24 keV

2+

1332

E = 1332.50 keV
0+

0
60

Fig. 3.4. Main part of the decay scheme of 60Co decaying by -decay
to 60Ni. On the right hand side of the states their energy referred to the
ground state is given (in keV), on the left hand side the nuclear spin and
parity characterising the respective level.

Ni

Quite often the radioactive decay modes as discussed above do not populate the ground states but
rather excited states of the daughter nuclides (cf. Fig. 3.4). These are de-excited mostly immediately by
one or more -transitions, until the ground state is reached. The energy of the -rays corresponds to the
energy difference of the nuclear levels involved in the respective transition.
Besides of -ray emission an excited nucleus can interact directly with a bound shell electron by
transfer of its energy leading to emission of the electron. This process - called internal conversion - is a
one-step process without the production of an intermediate -ray. It competes with the normal -emission,
preferentially for heavy nuclei, and obeys the same selection rules like -decay. The emission of an
electron of the K-shell is called K-conversion and analogous for the other shells L-, M-, N-conversion.
Spontaneous fission is a decay mode by which heavy nuclides with high neutron excess separate under
energy release into two lighter fragments and several neutrons. Among natural radioactive elements it
occurs only in very few cases for primordial or very long-lived nuclides heavier than uranium. Its
probability compared to the competing - or -decay is very small for uranium (238U: ~1:106), but
increases markedly with increasing atomic number Z and neutron excess. In addition, several short-lived
isomers decaying exclusively by spontaneous fission (fission isomers) have been observed for the
transuranium elements Np to Bk. Spontaneous fission is described by the transformation:
(Z, A)!(Z', A') + (ZZ', AA'x) + xn + E,
where (Z', A') and (ZZ', AA') denote the two fission fragments, x is the number of emitted neutrons and
E the energy released mainly in form of kinetic energy of the excited fragments and -radiation from
their de-excitation. More details on fission products and further physical background of fission will be
given in Chapter 3.6.
A further decay mode occurring, however, very rarely and mostly for nuclides far off the valley of
-stability is the emission of protons, neutrons or heavier nucleon clusters (5-7Li, 7-9Be, 11-14C, 14-16N, 19-22F
and 20-25Ne).
With increasing proton excess on the left-hand side of the valley of -stability, the binding energy of
the last proton decreases markedly, and proton emission from the ground state (p-decay) becomes
energetically possible by tunnelling through the energy barrier of the nuclear potential as in the case of
-decay. The emission of monoenergetic protons of 1.06 and 1.23 MeV, respectively, by transmutation of
the ground state of the mother nuclide into the ground state of the daughter nuclide was first observed for
147
Tm146Er + p (Ep = 1.051 MeV, t1/2 = 0.56 s) and 151Lu150Yb + p (Ep = 1.233 MeV, t1/2 = 85 ms).
Due to the competition with +-decay, which is favoured in most cases, p-decay from the ground state
Landolt-Brnstein
New Series VIII/4

3-18

3 Physical fundamentals

[Ref. p. 3-39

occurs very rarely. Much more frequently, p-emission occurs after +-decay feeding an excited state in
the daughter nuclide, from which the proton can easier surmount the energy barrier (+-delayed proton
emission). +-delayed emission of protons, in some cases even of two protons or an -particle, was found
for the lightest known isotopes of most of the elements between B and Zr.
The binding energy of additional neutrons is higher than that of additional protons and approaches
zero only at large distances from the line of -stability. Therefore, on the right-hand (neutron-rich) side of
the line of -stability all known nuclides are energetically stable to neutron emission from the ground
state, which has not been observed up to now. In contrast, neutron emission immediately following
-decay (-delayed neutron emission) is observed for many neutron-rich nuclides and many fission
products.
In very recent years spontaneous emission of particles heavier than -particles (cluster-emission) has
been observed in several cases. Spontaneous fragmentation of nuclei with atomic numbers Z >40 by
emission of cluster nuclei is energetically possible with extremely large partial half-lives. Consequently,
cluster radioactivity is a very rare event when compared to other decay modes.

3.4.3 The natural radioactive decay families


After the first detection of radioactivity in 1896 many other radioactive substances were identified in the
investigated uranium and thorium minerals. Among these, there are three primordial isotopes: 232Th, 238U
and 235U, which initiate three naturally occurring decay series. They were called the thorium, uraniumradium and actinium families, the latter two according to their most important members 226Ra and 227Ac,
respectively.
In these three decay series, only - and -decay occurs. By the emission of an -particle the mass
number decreases by 4 and the atomic number by 2 units, whereas by the emission of a -particle the
mass number remains unchanged and the atomic number increases by 1 unit. From this fact it follows that
all members of such a decay series have mass numbers differing from each other only by multiples of 4
units. This means that all mass numbers occurring in a series have mass numbers A = 4n + b, where n
varies within the series and b depends only on the mass number of the starting primordial isotope.
Consequently, the thorium family originating from 232Th (A = 232 = 4n with n = 58) is characterised by
the common label A = 4n; by variation of n, all possible mass numbers of the members of the decay series
are obtained. For the uranium-radium family starting with 238U, the respective label is A = 4n + 2, and for
the actinium family starting with 235U, A = 4n + 3. One radioactive decay series with A = 4n + 1 is
obviously missing in nature. Members of this family can be produced, however, artificially via nuclear
reactions. The longest half-life in this decay series belongs to 237Np; therefore it is called the neptunium
family. It was probably present in natural matter after the genesis of the elements before about 5 109 y
but decayed due to the relatively short half-life of 237Np.
The final members of all these decay series are of course stable nuclides: 208Pb at the end of the
thorium family, 206Pb for the uranium-radium family, 207Pb for the actinium family, and 209Bi for the
neptunium family. Furthermore, in all four decay series one or more branchings occur due to the
capability of several nuclides to decay by -decay as well as by -decay. In nearly all cases, however,
one branch preponderates strongly and the weak branch ends up at the same common stable end product
of the family.
In the early years of radioactivity only few radioactive elements and no concept of isotopy had been
established. Consequently, the newly detected activities, mainly distinguished from their differing halflives, were given historical names, like mesothorium, actinouranium, thoron, ..., according to the main
family members chemically identified at that time. These denominations are frequently found in older
literature. In Table 3.4 the abbreviations of these historical names are assigned to the respective isotopes.

Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-19

In the four diagrams in Fig. 3.5 all members of the 4 families are given together with their half-lives
and decay modes (arranged in a compressed form of the Segr type of charts of nuclides). For branching
decays the main decay branch is given first and indicated by a thick arrow pointing to the daughter
nuclide. A thin arrow marks a second weak branch (mostly < 1%) and is also used for subsequent decays
in these branches.
Table 3.3. Historical names (abbreviations) of the members of the three natural radioactive families
Thorium family A = 4n
232

Th
Th

228

228
Ra
Ac 228Th
MsTh1 MsTh2 RdTh

224

Ra
ThX

220

Rn
Tn

216

Po
ThA

212

Pb
ThB

212

Bi
ThC

212

Po
ThC'

208

Tl
ThC''

208

Pb
ThD

227

Th
RdAc
207
Tl
AcC''

223

Fr
AcK
207
Pb
AcD

223

Ra
AcX

219

219

Rn
An

215

215

Po
AcA

211

Pb
AcB

234

230

226

222

218

214

218

218

Actinium family A = 4n + 3
235

U
AcU
215
At

231

Th
UY
211
Bi
AcC

231

Pa

211
Po
AcC'

227

Ac

211m
Po

At

Bi

Uranium-radium family A = 4n + 2
238

U
UI
214
Bi
RaC

234

Th
UX1
214
Po
RaC'

Landolt-Brnstein
New Series VIII/4

234m

Pa

UX2
Tl
RaC''

210

234

Pa
UZ
210
Pb
RaD

U
UII
206
Hg

Th

Io
210

Bi
RaE

Ra

206
Tl
RaE''

Rn

210
Po
RaF

Po
RaA
206
Pb
RaG

Pb
RaB

At

Rn

3-20

3 Physical fundamentals

[Ref. p. 3-39

Fig. 3.5a-d (following). Decay chains of the 4 natural radioactive families in a compressed Segr type representation
(NZ over Z). In this representation -decay leads vertical to the second field below the mother nuclide, while
decay leads diagonal to the left field above. All chain members are given together with their half-lives and decay
modes. For branching decays the main decay branch is given first and indicated by a thick arrow pointing to the
daughter nuclide. A thin arrow marks a second mostly very weak branch and is also used for subsequent decays.

A = 4n

N-Z

44

46

48

50

52

Z
232

Th;
1.405.1010 y

228

Th;
1.913 y

90

228

Ac; 6.13 h

89
224

Ra;
3.66 d

88

228

Ra; 5.75 y

87
220

Rn;
55.6 s

86
85
84

212

Po;
0.15 s

212

Bi; , 60.60 m

83
82

216

Po;
300 ns

208

212

Pb; 10.64 h

Pb
stable

81

208

Tl; 3.053 m

(a) Thorium family

Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-21

A = 4n + 1

N-Z

43

45

47

49

51

53

Z
241

Am;
432.2 y

95

[238U(,n) ]
94

241

Pu; -,
14.35 y

237

Np;
2.144.106 y

93
233

U;
1.592.105 y

92

Pa; 27.0 d

229

Th;
7880 y

90

233

Th; 22.3 m

Ac;
10.0 d
225

Ra; 14.8 d

88
221

Fr;
4.9 m

87
217

Rn;
0.54 ms

86

217

At; , 32.3 ms

85
213

Po;
4.2 s

84

81

[(232Th+n) ]

225

89

82

U; 6.75 d

233

91

83

237

209

213

Bi; -,
45.59 m

Bi
stable
209

Pb; 3.253 h
209

Tl; 2.16 m

(b) Neptunium family


This decay chain is not directly initiated by a primordial nuclide but rather by nuclear reactions of the
naturally occurring nuclides 238U and 232Th, respectively, with -particles and fission neutrons from their
decay as indicated in the square brackets.
Landolt-Brnstein
New Series VIII/4

3-22

3 Physical fundamentals

[Ref. p. 3-39

A = 4n + 2

N-Z

42

44

46

48

50

52

54

Z
234

238

U;
2.455.105 y

92

U;
4.468.109 y
234g/m

Pa; - 6.7
h/1.17 m

91
230

Th;
7.54.104 y

90

234

Th; 24.10 d

89
226

Ra;
1600 y

88
87

222

Rn;
3.825 d

86
218

At;
2 s

85
84

210

81
80

218

Po; , 3.05 m

Po;
164 s

210

Bi; -,
5.013 d

83
82

214

Po;
138.38 d

206

214

Bi; -,
19.9 m

210

Pb; -,
22.3 y

Pb
stable
206

Tl; 4.2 m

214

Pb; 26.8 m

210

Tl; 1.3 m

206

Hg; 8.15 m

(c) Uranium-Radium family


In the Uranium-Radium family the decay of 234Th results in the population of the isomeric state (m) as
well as the groundstate (g) of 234Pa which in turn decay to 234U.

Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-23

A = 4n + 3

N-Z

43

45

47

49

51

Z
235

U;
7.038.108 y

92
231

Pa;
3.276.104 y

91
227

Ac; -,
21.773 y

223

Ra;
11.43 d

88

223

Fr; -,
21.8 m

87
219

Rn;
3.96 s

86
215

211

Po; , 1.78 ms

211

Bi; , 2.17 m

207

(d) Actinium family

215

Bi; 7.6 m

211

Pb; 36.1 m

Pb
stable

81

At; , 0.9 m

215

Po;
0.516 s

83

Landolt-Brnstein
New Series VIII/4

219

At;
0.1 ms

85

82

Th; 25.5 h

227

89

84

231

Th;
18.72 d

90

207

Tl; 4.77 m

3-24

3 Physical fundamentals

[Ref. p. 3-39

3.5 Radioactive radiation


3.5.1 Types of radiation
For the identification and investigation of radionuclides as well as for radiation protection the properties
and interactions of nuclear radiation have to be known. The most important aspect is the interaction of
radiation with matter, which determines the detection methods and the actions on inorganic and organic
matter.
Charged particles or photons, such as -particles, protons, electrons, positrons, - or X-rays, induce
ionisation processes in gases, liquids or solids and are called, therefore, ionising radiation. In addition to
the production of excited atoms or molecules also chemical reactions may be induced.
The minimum energy needed for ionisation or excitation of atoms or molecules is of the order of
several eV and depends on the type of the atoms or molecules involved. The photons of visible light and
neighbouring wavelenghts have energies between about 1 eV (wavelength = 1240 nm) and 10 eV ( =
124 nm). If their energy exceeds the ionisation energy of the absorbing matter, they lose it in a single
ionisation process. On the other hand, charged particles have energies in the range of 0.1 to 10 MeV and
produce a large number of ions, electrons and excited atoms or molecules in many interaction steps.
3.5.1.1 Particle radiation

This type of radiation comprises the frequently and also naturally occurring - and -radiation, the
neutron radiation accompanying mainly nuclear fission, and the quite rare types of nucleon- and clusteremission. The main properties of these particles influencing their physical behaviour like interaction with
and absorption in matter are their mass, charge and energy.
Ions and excited atoms or molecules produced in the primary interactions of charged particles with
matter give rise to further (secondary) physical and chemical reactions. Many of these secondary
reactions are very fast and happen relatively frequently. The concentration of such reaction products
along the path of the primary particle is proportional to its energy loss per unit path length. For example,
-particles of 1 MeV loose 190 eV/nm in water, whereas 1 MeV electrons loose only 0.2 eV/nm,
resulting in shorter ranges for the -particles but higher concentrations of reaction products, respectively.
Very high energetic particles may also induce nuclear reactions. Electrons loose in the force field of
nuclei part of their energy by generating bremsstrahlung. For electron energies of the order of 1 MeV
(e.g. -radiation), these photons have energies in the X-ray range and for energies >10 MeV the
bremsstrahlung photons have the energies of -rays.
Since neutrons carry no charge, they can interact with matter via the very short-range nuclear force,
and they loose their energy stepwise mainly by collisions with atomic nuclei or they may induce nuclear
reactions. This requires a large number of scattering processes and especially light materials to slow down
and absorb neutrons.
From range calculations it follows that -radiation is easily absorbed quantitatively (e.g. by a sheet of
paper), for absorption of -radiation several millimetres or centimetres of material are necessary, and for
absorption of -radiation thick layers of either lead or concrete are needed taking into account that the
absorption of -rays follows an exponential law. The ratio of the absorption coefficients for -, - and radiation of equal energy is about 104:102:1.

Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-25

3.5.1.2 Electromagnetic radiation

The types of electromagnetic radiation considered here are mostly produced during de-excitation
processes in the electron shells or the nuclei of atoms. Due to historical reasons, this electromagnetic
radiation despite of similar fundamental properties is differently denominated according to different
origins:

emission from de-exciting outer electron shells:


emission from de-exciting inner electron shells:
emission from de-exciting nuclei:
emission during deceleration of charged particles:

light
X-rays
-radiation
bremsstrahlung.

The energy range of X-rays lies within about 100 eV to 100 keV (= wavelengths of about 10 nm to
10 pm), and that of -rays within about 10 keV to 104 MeV (= wavelengths of about 0.1 nm to 107 nm).
That means there is an overlap in the energy ranges of X-rays and -rays. Electrons with energies
>10 MeV decelerated in a substance of high atomic number induce the emission of high-energetic
(hard) bremsstrahlung. In contrast to the -rays emitted from nuclei, this bremsstrahlung shows a
continuous energy distribution.
X-rays are emitted from the electron shell of the nuclides after formation of a hole in one of the inner
shells either by an external process (bombardment with photons, electrons or heavy charged particles) or
by an internal decay process like electron capture or internal conversion. The X-ray spectrum of an
individual nuclide shows a distinct line structure corresponding to the different transition possibilities in
the electronic shell and reflecting its structure.
The -rays emitted by an excited nucleus have well-defined energies, which correspond practically to
the differences in the excitation energies of the nuclei (the recoil energies transferred to the emitting
nuclei are very small). Gamma spectroscopy is, therefore, the usual method to investigate the level
structure and decay schemes of atomic nuclei.
Generally, the -radiation is emitted immediately after a preceding - or -decay, since the lifetimes
of excited states are of the order of 1013 s. However, if immediate -transitions are forbidden because
of high differences of the nuclear spins of the involved states and the ground state in combination with the
conservation laws of nuclear momentum and parity (selection rules), a metastable or isomeric state results
which decays with its own half-life different from that of the mother nuclide. The transition from the
isomeric to the ground state is called isomeric transition (IT). Some long-lived nuclear isomers in isotopes
with a stable ground state are of practical importance as pure -emitters.
3.5.1.3 Conversion electrons

If a -transition in an excited nucleus is hindered by the selection rules, a certain probability exists
increasing with increasing hindrance to transfer the excitation energy directly to a shell electron, which
is emitted monoenergetically. The energy Ee of these conversion electrons is given by Ee = E EB where
E is the respective -energy, and EB the binding energy of the electron in the respective shell. Since the
higher shells L, M, ... have sub-shells with slightly different binding energies, the conversion electron
spectra contain 3 lines for L-conversion and 5 lines for M-conversion.
The internal conversion leaves a hole in the electron shell, which by recombination causes the
emission of characteristic X-rays or Auger electrons. The latter in contrast to conversion electrons
gain their energy from transitions between electron shells, which is in general much lower than the
nuclear transition energies of conversion electrons.
The relative abundance of internal conversion is given by the total conversion coefficient = Ne/N,
which is composed from the partial conversion coefficients for the individual electron shells and subshells: = + L + LII + LIII + M..., each defined as the ratio of the number of the respective
conversion electrons to the number of -rays emitted.

Landolt-Brnstein
New Series VIII/4

3-26

3 Physical fundamentals

[Ref. p. 3-39

3.5.2 Physical properties of radiation


In order to investigate radionuclides and to develop protection methods against radioactive radiation, one
has to deal with their properties and the interaction processes between the different radiation types and
matter. In principle, the absorption of electromagnetic radiation like - and X-rays is different from that of
particles. While the latter lose their energy by successive collisions, photons give off their energy mostly
in one process. Because they are chargeless, their interaction with matter is small. The absorption of rays follows an exponential law:
I = I0 ed

(3.5.1)

where is the absorption coefficient and d the absorber thickness.


The exact validity of this exponential law is, however, restricted to monoenergetic -radiation, a
narrow beam of -rays and a thin absorber.
3.5.2.1 Interaction of charged particles with matter

The primary interactions of fast charged particles with matter can be classified as follows:
a)
b)
c)
d)
e)

elastic collisions with atomic electrons


inelastic collisions with atomic electrons
elastic collisions with nuclei
inelastic collisions with nuclei
nuclear reactions and interaction with nuclear forces

All these interactions can contribute in principle to deflection and deceleration of incoming particles.
Since more than 104 collisions are necessary to stop particles with energies in the MeV range, this is a
multi-scattering process composed statistically in different ways of the individual processes.
Heavy charged particles (p, d, , ions) loose most of their kinetic energy by process (b). The electrons
of the scattering atom are excited by the energy transfer, most of them to energies high enough to ionise
the atoms.
Elastic collisions of heavy particles (protons, deuterons, -particles, ...) with nuclei (c) are rare in
comparison to (b), for light particles (electrons, positrons), however, much more frequent. Inelastic
scattering from nuclei (d) also occurs rarely and is negligible for deceleration of heavy particles, for light
particles, however, it is important at higher energies giving rise to the emission of bremsstrahlung.
Consequently, there are characteristic differences for the deceleration of heavy and light particles. On
the other hand, the detection of all types of charged particles, as well as of electromagnetic radiation, is
based in general on their ionisation effects. This holds also for the damaging effects in biological
material.
Deceleration of heavy particles

By the inelastic scattering of fast particles from shell electrons the atoms are excited or preferentially
ionised. For the resulting loss of kinetic energy per unit path one obtains:

dE 4 z 2 e 4
=
N0B
ds
me v 2

(3.5.2)

In this expression also called stopping power z means the charge of the fast particle, v its velocity
and N0 the number of nuclei per cm3 in the absorber material.
Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-27

The atomic stopping number B is a function, depending on the energy of the incoming particle. From
a quantum mechanical calculation H. Bethe obtained the following expression for the atomic stopping
power B under consideration of relativistic effects at high particle energies:
2m v 2
C
B = Z ln e ln(1 2 ) 2 K
I
Z

(3.5.3)

where Z means the atomic number of the absorber, = v/c and I = 11.5Z the average ionisation potential;
CK is a correction factor depending on E and Z, which is of influence only at very low projectile energies
and takes values in between 0 and ~1.
For the stopping power it follows:

2m v 2
C
dE 4 z 2 e 4
=
N 0 Z ln e ln (1 2 ) 2 K
2
I
Z
ds
me v

(3.5.4)

The schematic course of equ. (3.5.4) shown in Fig. 3.6 demonstrates qualitatively that in the
intermediate region for energies much larger than the ionisation potential and much less than the particle
rest mass (I E Mc2) the energy loss decreases approximately as 1/E, since the logarithmic term varies
slowly and the relativistic terms are negligible. At high velocities, however, these terms give rise to a
slight increase causing a flat minimum at E 3Mc2. At low energies the logarithmic term prevails and the
curve decreases steeply below E 500I. From this behaviour it follows that the ionisation density
produced by the decelerated particle in the absorber increases markedly before the end of the path s and
decreases steeply behind this point. Furthermore, the 1/v2 dependence of the first term in equ. (3.5.4)
causes a stretching of the energy scale for different masses of the incoming projectiles (cf. Fig. 3.6).

Loss of kinetic energy


per unit path - d E
ds

Loss of kinetic energy


per unit path - d E [ MeV/ cm ]
dx

0.05

~
= 500 l

3 Mc 2
Particle energy E

0.04

0.03
e

0.02

0.01

0
10 -2

10 -1

1
10 2
10
Particle energy E [MeV]

10 3

10 4

Fig. 3.6. Schematic course of the stopping power with energy (left) and dependence on the projectile type for
electrons, muons, protons, deuterons and -particles (right).

3.5.2.2 Interaction of neutrons with matter

Neutrons are emitted by radioactive decay during spontaneous fission and in rare cases after -decay of a
very neutron-rich nuclide to an excited level of the daughter nuclide. In nuclear reactions and in particular
in nuclear fission neutrons are of high importance, especially for the production of radionuclides. Since
from radioactive sources mostly only small fluxes can be obtained, neutron sources based on nuclear
reactions, like 9Be(,n)12C, 9Be(d,n)10B or 9Be(,n)2, are used. The highest fluxes of neutrons are
available in nuclear reactors.
Landolt-Brnstein
New Series VIII/4

3-28

3 Physical fundamentals

[Ref. p. 3-39

The interaction of neutrons with matter happens mainly with nucleons and nuclei via elastic and
inelastic scattering and nuclear reactions, because they are electrically neutral and interact hardly with
electrons. Thus, ionisation by neutrons is negligible. In elastic collisions the total kinetic energy remains
constant and only deflection occurs, while in inelastic collisions part of the kinetic energy is used for
excitation of the collision partner. Depending on the neutron energy different types of interactions occur
and, correspondingly, several energy ranges are distinguished:

0-0.1 eV:
0.1-100 eV:
1-10 keV
0.1-100 keV:
0.1-10 MeV:

thermal neutrons (energies similar to those of gas molecules at room temperature)


slow neutrons
epithermal neutrons
neutrons of intermediate energies
fast neutrons

In contrast to charged particles, neutrons do not undergo Coulomb interaction with nuclei. However,
low-energy (thermal and slow) neutrons are very effectively absorbed by a great number of nuclei, giving
rise to nuclear reactions. Elements such as B, Cd, Sm, Eu, Gd and Dy are used as good neutron absorbers.
Epithermal neutrons are also called resonance neutrons, because there exist absorption maxima or
resonances at defined energies for distinct absorber nuclei. Neutrons with energies corresponding exactly
to the excitation energies of excited levels of the nucleus are absorbed with very high probability.
Fast neutrons are decelerated mainly by elastic and inelastic collisions. The energy released in one
elastic collision depends on the collision angle and the mass number of the target nucleus. The lighter the
nucleus, the higher is the energy loss of the neutron. Consequently, hydrogen or hydrogen-containing
substances like water or paraffin are very effective to reduce (moderate) the energy of neutrons.
Graphite can also be used as moderating material, but needs larger material thickness due to its higher
mass number. After deceleration the slow neutrons are captured by nuclei, giving rise to nuclear
reactions. High-energy (fast) neutrons may also induce nuclear reactions, but the probability and
consequently the contribution of this interaction type is relatively small.
3.5.2.3 Interaction of electromagnetic radiation with matter

Electromagnetic radiation passing a material experiences an intensity attenuation according to equ. (3.5.1)
(see above) via interactions with the various components of matter. For instance, -rays interact with:

atomic electrons,
nuclei,
electrical fields of the electrons and nuclei,
meson fields of the nuclei.

These interactions can result in energy losses, alterations of the propagation direction and polarisation.
The effects on -rays are:
total absorption,
inelastic scattering (incoherent),
elastic scattering (coherent, Thomson scattering).
Though all of the possible combinations of interactions and processes can occur, most of them result
in very weak effects, which can normally be neglected for attenuation considerations. Important are:
the photoeffect,
the Compton effect,
the pair formation.
In the photoeffect the incoming photon is totally absorbed by an atomic electron of the inner shells
(preferably of the K- or L-shells). A free electron cannot take up the total energy of the photon because of
the momentum conservation. In contrast, this is possible for the bound atomic electrons, because the atom
Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-29

takes up the residual momentum as recoil. Consequently, the stronger bound inner electrons exhibit the
strongest absorption. The photoelectrons in turn are emitted with a kinetic energy Ekin corresponding to
the total photon energy h reduced by the electron's binding energy Eb, which depends on the atomic
number of the respective atom:
Ekin = h Eb

(3.5.5)

Consequently, for high photon energies and light absorbers the photoelectrons carry nearly the
total photon energy. In addition, they exhibit a pronounced angular distribution favouring the forward
direction with increasing photon energy. Additional (isotropic) radiation is emitted originating from
the recombination process of the electron hole left by the emitted photoelectron. During this
recombination characteristic X-rays and Auger electrons are released by the energy set free by an
outer electron filling up the hole in the inner shell. Furthermore, bremsstrahlung is produced by the
photoelectrons especially in heavy materials. All these radiation types connected with the absorption
of photons together with the ionised residual absorber atoms can result in a high local energy density.
The Compton effect describes the inelastic scattering of photons from outer shell electrons, where the
photon looses only part of its energy, which is transferred to the electron. Both, the recoiling electron and
the photon are scattered with respect to the original photon direction, and the scattering into forward
directions is again favoured with increasing photon energy. The relative energy transfer to the electron
increases with increasing photon energy and also with increasing photon scattering angle. On the other
hand, the highest backscattering contributions occur at low photon energies.
The pair formation occurs in the electric field of an atomic nucleus, where the -ray can be
spontaneously transformed into an electron-positron pair, if its energy is larger than the sum of the rest
masses of the pair: E > 2mec2 = 1,022 MeV. The nucleus remains unchanged but is necessary for the
conservation of momentum. The positron annihilates by recombination with an electron of the absorber
atoms, whereby two annihilation -quanta are produced with an energy of 511 keV each.

Compton
effect

Total

Pair formation

Absorption coefficient m [cm -1]

Absorption coefficient m

Photoeffect
1.5

1.0
Pb

0.5
Cu
Fe

10 -1

1
10
- energy E [MeV ]

10 2

Fig. 3.7. Schematic representation of the partial and


total absorption coefficients of a heavy absorber in
dependence on the -ray energy.

0
0.1

Al
1
- energy E [MeV ]

10

Fig. 3.8.
Schematic representation of the total
absorption coefficients of different absorbers in
dependence on the -ray energy.

For the attenuation of photons in matter, mainly the three processes photoeffect, Compton effect and
pair formation have to be taken into account. Fig. 3.7 shows schematically their contributions to the total
absorption coefficient in dependence on the photon energy. A primary photon beam is attenuated by
scattering as well as by absorption. The main parameters for the attenuation are the photon energy and the
Landolt-Brnstein
New Series VIII/4

3-30

3 Physical fundamentals

[Ref. p. 3-39

atomic number and density of the absorber material (cf. Fig. 3.8). At low photon energies, classical
coherent (Thomson) scattering predominates, at higher energies incoherent scattering via the Compton
effect. Absorption of photon energy occurs mainly via the photoeffect and pair formation and partially via
the Compton effect. The lower energy threshold for the photoeffect is given by the binding energy of the
inner shell electrons, for the pair formation by the rest mass of the electron-positron pair (1022 keV). At
high atomic numbers Z and low photon energies the photoeffect dominates, at high energies the pair
formation. For energies between about 1 to 5 MeV the Compton effect dominates at all atomic numbers
Z. The resulting Compton electrons give, therefore, the most important contribution to the energy dose in
human tissue and to the biological effect of radiation.

3.6 Nuclear fission and fission products


3.6.1 Particle induced nuclear fission
Besides the few heavy nuclides decaying by spontaneous fission many other heavy isotopes undergo
fission after bombardment with particles, especially neutrons. Fission of uranium was first observed but
misinterpreted in 1934 by Fermi [34Fer] in an attempt to produce transuranium elements by irradiation of
uranium with slow neutrons. Similar experiments were performed by several groups, but only in 1937
Hahn, Meitner and Strassmann [37HMS] identified the observed radioactive products of the fission of
uranium to have appreciably lower atomic mass, such as 140Ba.
Fission of heavy nuclei always leads to products with a high neutron excess due to the much larger
neutron-to-proton ratio of heavy nuclides. The primary fission products are formed in about 1011 s by
fission and emission of prompt and -delayed neutrons and -rays from the highly excited fragments.
They always lie on the right hand side of the valley of -stability and decay by several successive
-decays following isobaric chains into nuclides of increasing atomic number Z ending up with the first
stable isobar in the chain.
The fission process exhibits different features depending mainly on the energy of the inducing
particles and on the atomic number Z of the fissioning nuclide. For fission induced by low-energy
neutrons with energies up to about 10 MeV two fission products with mass numbers in the range between
about 70 and 160 and 2-3 neutrons are emitted. The energy E released by fission is relatively high (E
200 MeV), since for the light fission products the binding energy per nucleon is higher than for the
heavy fissioning nuclei.
When comparing the naturally occurring uranium isotopes 234U (abundance: 0.0055 %), 235U (0.720
%), and 238U (99.2745 %), it is found that 235U undergoes fission by slow (i.e. thermal) neutron capture
with a remarkably large cross section of n,f = 586 1024 cm2. This holds also for other odd-mass (even
Z, odd N) nuclei like 233U, 235U, 239Pu and 241Pu. In all these cases the binding energy of an additional
neutron is very high resulting in high cross sections n,f for fission by slow (thermal) neutrons.
To induce fission in 238U, the neutron must have an energy of 1 MeV. The natural isotope 232Th
undergoes fission by 1.1 MeV neutrons. Using higher energetic particles, either neutrons, deuterons, or
-particles, it is possible to induce fission in any element with atomic number larger than 73.

3.6.2 Fission products


One of the most characteristic features of the fission process very important also for the activities that
have to be regarded in nuclear technologies is the resulting mass distribution of the various fission
products. As an example, Fig. 3.9 displays the yield of fission products found after thermal fission of 235U
Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-31

as a function of the mass number A. The maximum yields are around A = 90 to 100 and A = 133 to 143,
respectively. For these mass ranges the fission yields are about 6 %, whereas symmetrical fission around
A = 117 occurs with much smaller probability ( 0.01 %). It should be reminded that the sum of the
fission yields is 200 %, because each fission produces two fission products.
When investigating the mass distributions from fission with thermal neutrons one observes that those
of 233U and 239Pu are similar to that of 235U. In the case of 239Pu, however, the low mass maximum is
slightly shifted to higher masses, while the maximum for heavy fission products remains nearly
unchanged. This tendency continues with increasing mass of the fissioning nuclei, and in the case of
258
Fm the two maxima are superimposed.
An increase of the energy of the neutrons leads to a strong increase of the probability for symmetric
fission and gives rise to a flattening of the valley of the mass distribution by up to two orders of
magnitude. Increase of symmetric fission is also observed for nuclides with lower atomic numbers Z. For
227
Ac (Z = 89) symmetric and asymmetric fission have nearly the same probability, resulting in three
maxima in the mass distribution.
10

Fission yield [%]

10 -1

10 -2

10 -3

10 - 4
80

100

120
Mass number A

140

160

Fig. 3.9. Yields of fission products for the fission of


235
U by thermal neutrons.

It should be mentioned that the mass distribution curves as discussed before give the total yields of the
decay chains of mass numbers A. The independent yields for individual members of the decay chains, i.e.
the yields for direct formation in the fission process, are often difficult to determine, especially if in the
precursor chain short half-lives occur.
The total energy E released during fission consists of kinetic energy and excitation energy of the
primary fission fragments, where the kinetic energy resulting mainly from the Coulomb repulsion of the
two fission fragments gives the main contribution. In the case of low-energy fission the kinetic energy
Ekin is given by the empirical relation Ekin ~ Z2 / A1/3 where Z and A are the atomic number and the mass
number of the fissioning nucleus, respectively.
The primary fission fragments release their excitation energy by emission of prompt neutrons with
energies between 0 and about 10 MeV (mean value 2 MeV) and of prompt -rays. The number of
prompt neutrons emitted increases with the mass number of the fissioning nuclei and depends mainly on
the excitation energy of the primary fission fragments. Furthermore, an average number of 7.5 -rays with
a mean energy of about 1 MeV are emitted per fission, as well as several low-energy transitions in form
of conversion electrons and X-rays.
Landolt-Brnstein
New Series VIII/4

3-32

3 Physical fundamentals

[Ref. p. 3-39

In very rare cases high-energy charged particles such as p, d, t, -particles, 3He, 7Li, 8Li, 9Li, 9Be, 10Be
and isotopes of B, C, N and O are also emitted at an early stage of the low-energy fission process, when
the fission fragments are still very close to each other. The probability of this so-called ternary fission, i.e.
formation of three fragments increases strongly with the excitation energy of the fissioning nuclei. For
example, high-energy fission of 232Th with 400 MeV argon ions leads to a ratio of ternary to binary
fission of about 1:30.
In contrast to low-energy fission, high-energy fission induced by neutrons or other high-energy
particles leads to marked changes in the mass distribution of fission products. Among others, the
probability of symmetrical fission increases considerably with increasing excitation energy of the target
nuclei, resulting in a single flat maximum of the fission yield curve slightly below half the target mass
number.

3.6.3 Nuclear reactors


When comparing the energies available from combustion of carbon or carbon compounds (or in general:
from a chemical reaction like oxidation) with those from nuclear fission, one has to compare the orders of
magnitude of the binding energies in the electron shells and in the atomic nuclei, which are in the order of
eV and MeV, respectively. This means a difference of six orders of magnitude: 1 kg of carbon produces
an energy of 9.4 kWh, 1 kg of uranium can produce a maximum of 1.85 107 kWh. This high energy
release as already observed with the first fission experiments caused immediate considerations to make
use of this enormous energy potential. In addition, the neutrons released in each fission process could
initiate further fissions leading in principle to a continuous generation of energy. However, it soon turned
out that a safe control of a continuously proceeding fission process required the solution of various
technological questions and problems. Consequently, the first application of nuclear fission was the
construction of a nuclear explosive (atomic bomb) on the basis of uncontrolled self-amplifying fission
in the form of chain reactions.
As mentioned above, the fission process in the first step results in two fragments with high neutron
excess and in some prompt high energy neutrons. Furthermore, from the decay chain of the highly excited
primary fragments besides -particles and -rays about l % of -delayed neutrons are emitted which are
delayed by at least 0.01 second and about 0.07 % which are delayed by as much as 1 minute. This would
be a satisfactory condition for a chain reaction provided the neutrons originating from the fission process
have a possibility to react with other atoms of 235U. The neutrons generated during fission are, however,
no thermal neutrons but have energies up to the order of 1 MeV. If these neutrons are slowed down to
thermal velocities they can excite other 235U atoms to fission, so that the reaction would proceed with
increasing amplification as a chain reaction if only 235U would be present.
In natural uranium this process is not possible because of the high concentration of 238U atoms which
can capture higher energy neutrons without undergoing fission. In addition, there exist well-defined
energies, where a resonance absorption (in a very sharp energy window) of neutrons in 238U occurs with
very high cross sections (up to 1200 1024 cm2). Consequently, the fast neutrons have to be effectively
slowed down, whereby a sufficient number must escape the resonance absorption in 238U in order to reach
thermal velocities. This is achieved by use of a moderator, an element of small atomic weight like
deuterium, helium, beryllium, and carbon, whose atoms will not capture the neutrons but rather scatter
them elastically.

3.6.4 Nuclear explosives


The high amount of energy released by nuclear fission led very early to the production of nuclear
explosives. Since 235U, 233U and 239Pu have sufficiently high fission cross sections for fast neutrons, they
can be used as nuclear explosives if the respective critical masses are brought together. Without a neutron
Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-33

reflector increasing the number of neutrons within the explosive material, a sphere of about 50 kg
uranium metal containing 94 % 235U or a sphere of about 16 kg plutonium metal (239Pu) reaches
criticality. If a neutron reflector is used, the critical masses are about 20 kg for 235U and about 6 kg for
239
Pu. The critical masses for 233U are similar to those for 239Pu.
The use of plutonium in nuclear weapons requires a low concentration of 240Pu in the plutonium,
because its presence leads to the production of high numbers of neutrons by spontaneous fission.
Consequently; a too high concentration of 240Pu would initiate the neutron multiplication too early with a
relatively small multiplication factor and a relatively low energy release. Higher concentrations of 241Pu
also interfere, because of its decay into 24lAm with a half-life of only 14.4 y. To minimise the formation
of 240Pu and 241Pu, Pu for use in weapons is, in general, produced in special reactors with low burn-up
( 20000 MWthd per ton).
Criticality can be reached by shooting two under-critical hemispheres onto each other by means of
normal explosives (gun-type) or by compressing an under-critical spherical shell into a supercritical
sphere (implosion-type). The bomb ignited over Hiroshima (energy release corresponding to 15 kilotons
of TNT) was of the gun type using 235U, whereas that ignited over Nagasaki was of the implosion type
using 239Pu (energy release corresponding to 22 kilotons of TNT). Generally, the fissile core is
surrounded by a heavy material, in order to reflect the neutrons and to increase the inert mass and
consequently the time in which the super-critical configuration is held together.
The explosion of fissile material leads to temperatures of about 108 K which are sufficient to initiate
fusion between deuterium and tritium. This is the basis of the development of hydrogen bombs, in which
the energy of fission is used for ignition of fusion. LiD serves as a source of D and T, the latter being
produced by thermal (6Li(n,n)d) and by fast neutrons (7Li(n,n)t). If the temperature is high enough, the
D-D reaction can contribute to the energy production. The fast neutrons released by the fusion reactions
react very effectively with natural or depleted U initiating fission of 238U. By these kinds of weapons large
amounts of fission products are formed (dirty weapons). If a surrounding of non-fissile heavy material
is used, fission products are released only by the ignition process (clean weapons).

3.6.5 Radioactive inventory and nuclear waste


The radioactive inventory in a nuclear power reactor originates from:
fission products
uranium and transuranium elements formed by direct neutron induced reactions and their decay chains
isotopes produced by nuclear reactions in the cladding material of fuel rods, the reactor vessel
components and the coolant.
The dominating longer-lived fission products occurring in spent fuel elements are 85Kr, 131I, 133I,
Xe, 135Xe, 134Cs and 137Cs. Radionuclides produced by nuclear reactions in the coolant are 3H, 14C, 13N,
16
N, 19O, 18F and 41Ar. Furthermore, fission products or actinides may leak into the cooling system from
faulty fuel elements. Other radionuclides are produced by reactions with metals and their corrosion
products in various reactor vessel components like 51Cr, 54Mn, 59Fe, 58Co, 60Co, 65Zn, 124Sb.
133

Landolt-Brnstein
New Series VIII/4

3-34

3 Physical fundamentals

[Ref. p. 3-39

10 3

Rel.activity

a
10 2
b

Fig. 3.10. Radioactive decay of the sum of all fission


products (a), 106Ru (0.5 %, T = 373.6 d) (b), and 137Cs
( 6 %, T = 30.17 y) (c).

10
c
5
0

100 200 300 400 500 600 700 800 900


Decay time [d ]

Shortly after shut-down of a nuclear reactor the activity of the fuel is 1.7 1017 Bq per MW of
thermal energy produced. 237U (from the reactions 235U(n, )236U(n, )237U and 238U(n, 2n)237U) causes a
relatively high initial uranium activity. Since it decays with a half-life of 6.75 d, it vanishes rapidly after
the necessary storage of the discharged fuel elements. A global composition of spent nuclear fuel from
light water reactors after storage of 1 year is given in Table 3.5.
The radioactive waste produced during the operation of nuclear reactors is usually classified according
to the state of matter (gaseous, liquid or solid) and according to the activity level as low-active waste
(LAW), medium-active waste (MAW), and high-active waste (HAW). The largest amount of
radioactivity is concentrated in the spent fuel elements representing highly-active waste (HAW).
Table 3.4. Main components of spent nuclear fuel from a light-water reactor with an initial enrichment of
3.3 % 235U, a burn-up of 34000 MWd per ton and a storage time of 1 year (from [97Lie]).

Nuclide

Weight percent

uranium and transuranium elements


235
U
0.756
236
U
0.458
237
U
3109
238
U
94.2
237
Np
0.05
238
Pu
0.018
239
Pu
0.527
240
Pu
0.220
241
Pu
0.105
242
Pu
0.038
americium isotopes
0.015
curium isotopes
0.007

Nuclide

Weight percent

fission products
85
K
90
Sr
129
I
134
Cs + 137Cs
others
Stable fission products

0.038
0.028
0.09
0.275
0.19
3.0

Reprocessing of nuclear fuel transforms all waste types into liquid solutions and results in the
following amounts per ton of U: 1 m3 HAW (fission products and actinides in HNO3 solution), 3 m3
MAW as organic solution, 17 m3 MAW as aqueous solution, 90 m3 LAW (aqueous solution). By
further processing a volume reduction is achieved: ; 0.1 m3 HAW, 0.2 m3 MAW (organic), 8 m3
MAW (aqueous), 3m3 LAW (aqueous). After respective storage times the HAW solutions are
transformed by calcination or vitrification into stable forms like ceramics or glasses suitable for long-term
disposal and also in order to reduce the volume.
Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-35

After one year of intermediate storage and reprocessing, the initial activity of the HAW solutions is of
the order of 1014 Bq/l from which the activity due to 90Sr and 137Cs is about 1013 Bq/l; after 10 y the
activity of the HAW solution decreases approximately with the half-life of these nuclides (28.64 y and
30.17 y, respectively, cf. Fig. 3.10). After 1000 y the residual activity (of the order of 104 Bq/l) is
determined by long-lived fission products like 99Tc, 129I and actinides.
Solid MAW and LAW originate from structure material of the fuel elements, undissolved, dispersed
and filtered particles of metals or metal oxides (e.g. Ru, Rh, Mo, Tc), and gaseous components like
tritium (as T2 or HTO), 14C (as CO2), 85Kr, 129I or 106Ru (as RuO4) after adsorption in special adsorbents.
Computer codes have been developed to calculate the inventory of all radionuclides in spent fuel rods
after discharge and after several periods of storage. As an example in the Tables 3.6 and 3.7, taken from
[95SSK], the activities of actinides, fission products and light elements from surrounding material are
listed, which were obtained from a calculation with the code KORIGEN for a given reactor type, initial
235
U enrichment, and fuel burn-up at charge time, discharge time and several storage times. Further
information on these calculations and their results as well as on general reprocessing, recycling and
disposal concepts can be found e.g. in [83FiW], [93Wie], [95SSK], [80Clo] and [97Lie].
Table 3.5. Inventory of selected relevant radionuclides in Bq for a pressurized water reactor with a
thermal power of 3.733 MW after an operation time of 1 day [95SSK].

Nuclide
85

Time after discharge


1h
6h

0h

24 h

120 h

Kr
Kr
87
Kr
88
Kr
133
Xe
135
Xe
Kr-Xe

13

4.0810
1.431018
2.861018
4.031018
2.891017
1.741018
1.031019

13

4.2910
1.241018
1.681018
3.161018
3.101017
2.091018
8.471018

13

4.9610
5.721017
1.101017
9.321017
4.041017
2.911018
4.931018

13

5.5010
3.531016
6.021012
1.151016
6.151017
1.721018
2.381018

5.531013
1.251010
0.0
7.601005
5.821017
2.041015
5.841017

131

I
I
133
I
134
I
135
I
Iodine

2.361017
9.661017
4.161018
8.761018
6.761018
2.091019

2.431017
9.611017
4.151018
6.531018
6.091018
1.801019

2.461017
9.261017
3.571018
2.631017
3.601018
8.611018

2.381017
7.921017
1.961018
2.231011
5.461017
3.541018

1.801017
3.381017
8.011016
0.0
2.321013
5.981017

89

7.291016
4.301014
5.511018
1.081014
4.111016
7.871016
4.281018
7.681014
1.581018
9.381017
6.801016
1.401018
1.221015
4.101017
2.271016
7.541017

7.421016
4.321014
5.141018
1.121014
4.371016
7.951016
4.111018
8.321014
l.561018
9.861017
6.811016
1.241018
1.221015
4.281017
2.321016
6.531017

7.411016
4.311014
3.571018
1.291014
5.471016
7.931016
3.351018
1.151015
1.481018
1.141018
6.781016
5.671017
1.221015
4.681017
2.361016
2.931017

7.331016
4.311014
9.591017
1.831014
7.281016
7.871016
1.601018
2.291015
1.231018
1.151018
6.691016
3.411016
1.221015
3.811017
2.091016
1.631016

6.941016
4.311014
8.711014
3.441014
7.601016
7.531016
3.121016
7.931015
4.481017
4.321017
6.241016
1.051010
1.211015
5.881016
1.021016
3.321009

85rn

132

Sr
Sr
91
Sr
90
Y
91
Y
95
Zr
97
Zr
95
Nb
99
Mo
99m
Tc
103
Ru
105
Ru
106
Ru
105
Rh
127
Sb
129
Sb
90

Landolt-Brnstein
New Series VIII/4

3-36

3 Physical fundamentals

[Ref. p. 3-39

Nuclide
127
Te
127rn
Te
129
Te
129m
Te
131m
Te
132
Te
134
Cs
136
Cs
137
Cs
140
Ba
140
La
141
Ce
143
Ce
144
Ce
143
Pr
239
Np
238
Pu
239
Pu
240
Pu
241
Pu
241
Am
242
Cm
244
Cm
Aerosols

0h
1.071016
1.071013
6.701017
1.791015
1.721017
9.481017
3.671010
2.991014
4.561014
3.831017
7.201016
1.071017
2.661018
1.521016
7.231016
1.501019
8.381006
6.181011
5.251009
3.601009
3.871003
2.151002
9.621004
3.521019

Time after discharge


1h
6h
1.131016
1.411016
1.161013
1.591013
17
6.4310
3.421017
15
1.8710
2.111015
17
1.7010
1.521017
17
9.4210
9.011017
10
3.7710
4.021010
14
2.9810
2.951014
14
4.5810
4.581014
3.821017
3.781017
16
7.7310
1.021017
17
1.1310
1.301017
18
2.6510
2.391018
16
1.5210
1.521016
16
7.7810
1.041017
19
1.5110
1.431019
06
9.4210
1.441007
11
6.6710
9.091011
09
5.5110
5.791009
09
3.6010
3.601009
03
4.5310
7.821003
02
2.6710
4.951002
03
1.2610
2.461003
19
3.4610
3.001019

24 h
1.771016
3.061013
2.081016
2.271015
1.001017
7.681017
4.131010
2.831014
4.581014
3.631017
1.741017
1.401017
1.631018
1.521016
1.741017
1.151019
2.991007
1.671012
5.811009
3.601009
1.971004
1.011003
4.551003
2.051019

120 h
9.851015
8.181013
1.371015
2.101015
1.091016
3.281017
4.121010
2.291014
4.581014
2.921017
2.901017
1.291017
2.181017
1.501016
2.681017
3.531018
7.041007
3.791012
5.811009
3.601009
8.281004
1.411003
5.401003
6.371018

Total

6.271020

1.461020

3.291019

7.961018

7.091019

Table 3.6. Inventory of selected relevant radionuclides in Bq for a pressurized water reactor with a
thermal power of 3.733 MW after an operation time of 333 days [95SSK]

Nuclide
85
Kr
85m
Kr
87
Kr
88
Kr
133
Xe
135
Xe
Kr-Xe

0h
1.611016
1.191018
2.271018
3.221018
7.621018
1.831018
1.611019

Time after discharge


1h
6h
1.611016
1.611016
1.041018
4.781017
18
1.3310
8.721016
18
2.5310
7.451017
18
7.6210
7.601018
2.201018
3.071018
19
1.4710
1.201019

24 h
1.611016
2.951016
4.781012
9.191015
7.351018
1.821018
9.221018

120 h
1.611016
1.051010
0.0
6.081005
4.751018
2.161015
4.771018

131

3.501018
5.181018
7.631018
8.381018
7.141018
3.181019

3.501018
5.161018
7.501018
6.001018
6.431018
2.861019

3.251018
4.271018
3.521018
1.961011
5.771017
1.161019

2.331018
1.821018
1.431017
0.0
2.451013
4.301018

I
I
133
I
134
I
135
I
Iodine
132

3.451018
4.991018
6.401018
2.331017
3.811018
1.891019

Landolt-Brnstein
New Series VIII/4

Ref. p. 3-39]

3 Physical fundamentals

3-37

Nuclide
89
Sr
90
Sr
91
Sr
90
Y
91
Y
95
Zr
97
Zr
95
Nb
99
Mo
99m
Tc
103
Ru
105
Ru
106
Ru
105
Rh
127
Sb
129
Sb
127
Te
127m
Te
129
Te
129m
Te
131m
Te
132
Te
134
Cs
136
Cs
137
Cs
140
Ba
140
La
141
Ce
143
Ce
144
Ce
143
Pr
239
Np
238
Pu
239
Pu
240
Pu
241
Pu
241
Am
242
Cm
244
Cm
Aerosols

0h
4.491018
1.261017
5.421018
1.301017
5.581018
6.701018
6.521018
6.551018
6.911018
6.051018
4.881018
2.801018
6.571017
2.661018
2.581017
1.011018
2.501017
2.801016
9.891017
1.471017
4.861017
5.121018
8.261016
6.901016
1.511017
6.871018
6.951018
6.451018
6.181018
3.251018
6.101018
6.251019
5.761014
8.831014
5.911014
1.101017
4.161013
4.731015
2.921013
1.671020

Time after discharge


1h
6h
4.481018
4.471018
1.261017
1.261017
18
5.0510
3.501018
17
1.3010
1.301017
18
5.5710
5.571018
18
6.7010
6.681018
18
6.2610
5.101018
18
6.5510
6.551018
18
6.8410
6.491018
6.051018
5.951018
18
4.8810
4.861018
18
2.4710
1.131018
17
6.5710
6.571017
18
2.6610
2.571018
17
2.5710
2.501017
17
8.6910
3.891017
17
2.5010
2.491017
16
2.8010
2.801016
17
9.5210
5.491017
17
1.4710
1.461017
17
4.7710
4.251017
18
5.0810
4.861018
16
8.2610
8.261016
16
6.8910
6.811016
1.511017
1.511017
18
6.8610
6.781018
18
6.9410
6.931018
18
6.4510
6.441018
18
6.0910
5.491018
18
3.2510
3.251018
18
6.1010
6.101018
19
6.2110
5.851019
14
5.7610
5.771014
14
8.8410
8.851014
14
5.9110
5.911014
17
1.1010
1.101017
13
4.1610
4.171013
15
4.7410
4.751015
13
2.9210
2.931013
20
1.6510
1.551020

24 h
4.431018
1.261017
9.421017
1.291017
5.541018
6.631018
2.441018
6.551018
5.371018
5.141018
4.801018
6.821016
6.561017
1.911018
2.191017
2.171016
2.321017
2.801016
1.201017
1.441017
2.811017
4.141018
8.251016
6.551016
1.511017
6.511018
6.851018
6.351018
3.761018
3.251018
6.041018
4.691019
5.801014
8.881014
5.911014
1.101017
4.211013
4.761015
2.941013
1.301020

120 h
4.191018
1.261017
8.551014
1.271017
5.291018
6.351018
4.751016
6.551018
1.961018
1.891018
4.471018
2.101010
6.511017
2.931017
1.071017
4.421009
1.301017
2.791016
8.651016
1.331017
3.051016
1.771018
8.221016
5.301016
1.511017
5.241018
5.911018
5.831018
5.011017
3.211018
5.211018
1.451019
5.881014
8.961014
5.911014
1.101017
4.401013
4.711015
2.951013
7.501019

Total

7.751020

3.121020

1.751020

9.691019

Landolt-Brnstein
New Series VIII/4

2.331020

3-38

3 Physical fundamentals

[Ref. p. 3-39

3.6.6 Release of radionuclides from the radioactive inventory of a nuclear


reactor
In case of an accident of a less dangerous category mainly gaseous or volatile fission or decay products
are released. Heavier elements or compounds are more or less retained, since they are hardly transported
over longer distances. Consequently, the respective risk assessment studies show the highest risk
potentials for the gaseous fission or decay products like: noble gases, iodine, radon, volatile elements and
compounds (cf. e.g. [95SSK]).
Furthermore, it has to be taken into account that the chemical and physical behaviour of many fission
products may change considerably after nuclear transformations within their radioactive decay chains
(e.g.: Ra Rn). This often leads to consequences for the retention behaviour, release of and filter
effectiveness for various nuclides in the radioactive inventory.

Landolt-Brnstein
New Series VIII/4

3 Physical fundamentals

3-39

3.7 References
10Bat
34Fer
37HMS
79Ewb
80Clo
83FiW
93Wie
95AMC
95SSK

96Bec
97Lie
98Cur

98PKS
98Sch
99Mag
99MoT

Bateman, H.: Proc. Cambridge. Philos. Soc. 15 (1910) 423.


Fermi, E.: Nature (London) 133 (1934) 898.
Hahn, O., Meitner, L., Strassmann, F.: Z. Phys. 106 (1937) 249.
Ewbank, W.B., Ellis, Y.A., Scmorak, M.R.: Nucl. Data Sheets 26 (1979) 1.
Closs, K.D. (ed.): Report KfK 3000, 1980.
Fischer, U., Wiese, H.W.: Report KfK 3014, 1983.
Wiese, H.W.: Nucl. Technol. 102 (1993) 68.
Adloff, J.P., MacCordick, H.J.: Radiochim. Acta 70/71 (1995) 13.
Leitfaden fr den Fachberater Strahlenschutz der Katastrophenschutzleitung bei kerntechnischen
Notfllen, Verffentlichungen der Strahlenschutzkommission Band 13; Herausgegeben vom
Bundesministerium fr Umwelt, Naturschutz und Reaktorsicherheit; 2. berarbeitete Auflage,
Stuttgart, Jena, New York: Gustav Fischer Verlag, 1995.
Becquerel, H.: C. R. Seances Acad. Sci. (Paris) 122 (1896) 501.
Lieser, K.H.: Nuclear and radiochemistry: Fundamentals and applications, D-69451
Weinheim (Federal Republic of Germany): VCH Verlagsgesellschaft mbH, 1997.
Sklodowska-Curie, M.: C. R. Seances Acad. Sci. (Paris) 126 (1898) 1101.
Curie, P, Sklodowska-Curie, M.: C. R. Seances Acad. Sci. (Paris) 127 (1898) 175.
Curie, P., Curie, M., Bmont, G.: C. R. Seances Acad. Sci. (Paris) 127 (1898) 1215.
Sklodowska-Curie, M.: Rev. Gn. Sci. Pures Appl. Bull. Assoc. Fr. Av. Sci. 10 (1899) 41.
Curie, P., Curie, M., Bmont, G.: Sci. Am. 80 (1899) 60.
Curie, P., Curie, S.: C. R. Seances Acad. Sci. (Paris) 134 (1902) 85.
Pfennig, G., Klewe-Nebenius, H., Seelmann-Eggebert, W.: Chart of the nuclides (Karlsruher
Nuklidkarte), 6th ed., reprint 1998, Forschungszentrum Karlsruhe: Technik und Umwelt, 1998.
Schmidt, G.C.: Verh. Dtsch. Phys. Ges. 17 (1898) 14; C. R. Seances Acad. Sci. Paris 126
(1898) 1264.
Magill, J.: Nuclides 2000: An electronic chart of the nuclides; EUR 18737 EN, 1999.
Mohr, P.J., Taylor, B.N.: J. Phys. Chem. Ref. Data 28 (1999) 1713.

Landolt-Brnstein
New Series VIII/4

Ref. p. 4-27]

4 Radiological quantities and units

4-1

4 Radiological quantities and units

In this Chapter the fundamental quantities and units for ionising radiation and in addition specific
quantities used in radiological protection are described.

4.1 Introduction
While radiation field quantities, quantities describing radioactivity and absorbed dose quantities are based
on physical phenomena only, specific dose quantities in radiation protection as e.g. effective dose, include
factors which are based on judgements about the biological response of tissues, e.g. due to cancer
induction. These factors have been changed in the past in view of new research results and ideas. The
definitions given are mainly based on ICRU Report 51 [93I1], ICRU Report 60 [98I1], ICRP Publication
60 [91I1] and the ISO Standards Handbook, Quantities and Units [93I2].

Stochastic and non-stochastic quantities


Physical quantities are used to describe physical phenomena or objects. Many physical processes, e. g. the
decay of radionuclides, the number of interactions in a small volume irradiated or the energy transferred,
are subject to inherent fluctuations. This situation is described by stochastic quantities the values of
which follow a probability distribution. Some times this may be a Poisson distribution which is uniquely
determined by its mean value. In many other cases a quantity is defined by averaging in time or over a
volume which results in a single value with no inherent fluctuation. Those quantities, e. g. fluence or
absorbed dose, are called non-stochastic quantities.

Units
A unit is a reference sample of a quantity with which other quantities of the same kind are compared.
Every quantity is expressed as a product of a numerical value and a unit. Generally the use of the
International System of Units (SI) as given by the BIPM [91BI] is recommended which is based on the 7
base units meter, kilogram, second, ampere, kelvin, mole and candela. Derived SI-units are often given
special names like joule, becquerel or gray. Some other units are, however, generally used which are
outside of the international system, e. g. the electron volt (eV) and the atomic mass unit (u) - and the time
units minute, hour, day and year are also generally permitted.
Nevertheless, other units even if not recommended are still in use in radiation measurements and
radiation protection. Table 1 presents some numerical relationships between those units and the SI-units
recommended.
Landolt-Brnstein
New Series VIII/4

4-2

4 Radiological quantities and units

[Ref. p. 4-27

Table 4.1. Former units and its relations to SI-units


Quantity

Symbol

SI-unit Name

Former units
rad

1 rad = 0.01 Gy

roentgen

1 R = 2.58 104 J kg1

rem

1 rem = 0.01 Sv

absorbed dose

J kg1

exposure

C kg1

dose equivalent

J kg1

sievert (Sv)

activity

s1

becquerel (Bq) curie

potential alpha energy

cp

J m3

gray (Gy)

concentration
potential alpha energy
exposure

1 Ci = 3.7 1010 Bq

Working level 1 WL = 2.08 105 J m3


= 1.30 108 MeV m3

Ep

J h m3

Working level month (T = 170 h)


1 WLM = 3.54 103 J h m3
= 2.21 1010 MeV h m3

4.2 Radiation field quantities


Radiation field quantities are non-stochastic quantities defined at any point of a radiation field. Radiation
fields may consist of various types of particles and the field quantities are always related to a specific
particle type. This is usually expressed by adding the particle name to the quantity, e.g. photon fluence or
neutron flux. There are two classes of radiation field quantities referring either to the number of particles
or to the energy transported by them.
A radiation field of a specific particle type can be fully described by the number N of particles, their
distribution in energy as well as their spatial, directional and temporal distribution. This needs the
definition of scalar and vectorial quantities. While in radiation dosimetry mostly scalar field quantities are
used, vectorial quantities are often needed and applied in radiation transport theory and calculations. The
radiation field quantities are defined in specifying the field in increasing detail.

4.2.1 Scalar radiation field quantities


Particle number, radiant energy
The particle number, N, is the number of
particles that are emitted, transferred, or
received.
Unit: 1

The radiant energy, R, is the energy (excluding rest energy) of the particles that are
emitted, transferred, or received.
Unit: joule, J

Landolt-Brnstein
New Series VIII/4

Ref. p. 4-27]

4 Radiological quantities and units

4-3

For particles of energy E (excluding rest energy), the radiant energy, R, is equal to NE.
The distributions, NE and RE, of the particle number and the radiant energy with respect to energy are
given by
NE = dN/dE

and RE = dR/dE

(4.2.1a+b)

where dN is the number of particles with energy between E and E + dE and dR is their radiant energy.
Flux, energy flux
The flux, N& , is the quotient of dN by dt, where
dN is the increment of the particle number in
the time interval dt.
N& = dN/dt
Unit: s1

The energy flux, R& , is the quotient of dR by


dt, where dR is the increment of the radiant
energy in the time interval dt.
R& = dR/dt
Unit: W

The term flux has often been employed for the quantity fluence rate (see below). This usage should be
avoided.
Fluence, energy fluence
The quantity fluence is based on the idea of counting the number of particles incident or passing a small
sphere. It is defined by:
The fluence, , is the quotient of dN by da,
where dN is the number of particles incident on
a sphere of cross-sectional area da.
= dN /da
Unit: m2

The energy fluence, , is the quotient of dR


by da, where dR is the radiant energy incident
on a sphere of cross-sectional area da.
= dR /da
Unit: J m2

The fluence is independent of the directional distribution of the particles passing the sphere. In
calculations, fluence is often expressed in terms of the length of trajectories of particles passing a volume
dV. The fluence, , is given by

= dl /dV

(4.2.2)

where dl is the sum of the lengths of trajectories through this volume.


The distributions, E and E, of the fluence and energy fluence with respect to energy are given by

E = d/dE

and E = d/dE

(4.2.3a+b)

These quantities are often called spectral fluence and spectral energy fluence, respectively.
Fluence rate, energy fluence rate
The temporal distribution of the fluence and energy fluence is generally of interest. This results in the
following definitions:
The fluence rate, & , is the quotient of d by
dt, where d is the increment of the fluence in
the time interval dt.
& = d /dt
Unit: m2 s1

Landolt-Brnstein
New Series VIII/4

The energy fluence rate, & , is the quotient


of d by dt, where d is the increment of the
energy fluence in the time interval dt.
& = d /dt
Unit: J m2

4-4

4 Radiological quantities and units

[Ref. p. 4-27

The fluence rate has often been termed particle flux density. Because the term density mostly
characterises a mass density (kg1), it is recommended to use the term fluence rate and not particle flux
density.
Particle radiance, energy radiance
The energy radiance, & , is the quotient of
d & by d, where d& is the energy fluence
rate of particles propagating within a solid
angle d around a specified direction.
& = d& / d
Unit: W m2 sr1

The particle radiance, & , is the quotient of


d & by d, where d & is the fluence rate of
particles propagating within a solid angle d
around a specified direction.
& = d& / d
Unit: m2 s1 sr1

The specification of a direction requires two variables. In a spherical coordinate system with a
polar angle, , and an azimuthal angle, , d is equal to sin d d.
The distribution of particle radiance and energy radiance with respect to energy are given by

& ,E = d& / d dE and & ,E = d& / d dE

(4.2.4a+b)

4.2.2 Vectorial radiation field quantities


Radiometric quantities are often used to describe the flow of radiation in specific directions. This needs
the definition of vectorial quantities. For example, the scalar angular differential quantities like particle
radiance and energy radiance are transferred to vectorial quantities by multiplication with the unit vector
in a specific direction. Vectorial quantities are
vectorial particle radiance, &

with

vectorial energy radiance, &


vectorial fluence rate, &

with

vectorial energy fluence rate, &

with

vectorial fluence,

with

vectorial energy fluence,

with

with

& = &

&
&
=
& = & d

&
= & d
= & dt
= & dt

unit: m2 s1 sr1,
unit: W m2 sr1,
unit: m2 s1,
unit: W m2,
unit: m2,
unit: J m2.

A detailed description is given in ICRU Report 60 [98I1]. The distribution of a quantity with respect
to energy of the particle considered is described by an index E similar to the scalar quantities. For
example, the distribution of the vectorial particle radiance is given by
& ,E = & ,E
unit: m2 s1 sr1 MeV1.

Landolt-Brnstein
New Series VIII/4

Ref. p. 4-27]

4 Radiological quantities and units

4-5

4.3 Interaction coefficients and quantities


Ionising radiation is either charged (e.g. electrons, positrons, protons and -particles) or uncharged (e.g.
photons and neutrons). This dominates the main interaction with matter. While charged particles (called
directly ionising particles) are mainly slowed down by electromagnetic interactions with electrons of the
target atoms, the uncharged particles (indirectly ionising particles) interact with matter in separated
events. Indirectly ionising particles are either absorbed or its energy and direction is altered.
The probabilities of specific interactions between radiation and matter are characterized by interaction
coefficients. They refer to specific interaction processes, type and energy of radiation and the matter
involved. The definition of those coefficients important for dosimetry and related quantities are given in
this Section.

4.3.1 Cross section


The cross section is the most fundamental interaction coefficient. It is defined as follows.
The cross section, , of a target entity, for a particular interaction produced by incident particles is the
quotient of P by , where P is the probability of that interaction for a single target entity when subjected
to the particle fluence, . It is

= P/

unit: m2.

A special unit often used for the cross section is the barn (b) with 1 b = 1028 m2.
Cross sections mostly vary with the energy of the incident radiation (notation: (E)). The distribution
of a cross section with respect to the energy and direction of the emitted radiation is often called
differential cross section (d/d : angular differential cross section, d/dE: energy differential cross
section, d2/dEd : energy and angular differential cross section).
The total cross section, T, is the sum of the cross sections of all possible interaction channels for an
incident particle of a given type and energy and a given target material.

4.3.2 Mass attenuation coefficient and mass energy transfer coefficient


For an infinite small parallel beam of uncharged radiation, the interaction of radiation with matter results
in an attenuation of the incident beam with depth in material. This can be described by the relation
dN
= dl
N

(4.3.1)

where dN/N is the fraction of particles that experience interactions in traversing a distance dl in the
material. is the linear attenuation coefficient. The reciprocal of is called the mean free path of an
uncharged particle. In first order is proportional to the density of a material. This leads to the
definition of the mass attenuation coefficient, / .

1 dN
=
dl N

Landolt-Brnstein
New Series VIII/4

unit: m2 kg1

(4.3.2)

4-6

4 Radiological quantities and units

[Ref. p. 4-27

The mass attenuation coefficient is related to the total cross section by


N
NA
=
= A
M T M

(4.3.3)

where NA is the Avogadro constant and M the molar mass of the material considered. J are the cross
section related to the interaction of type J in this material.
For uncharged particles the transfer of energy to charged particles in the material is of high interest in
dosimetry. This is expressed by the mass energy transfer coefficient, tr/ , which is defined by
tr
1 dRtr
=

dl R

unit: m2 kg1

(4.3.4)

where dRtr/R is the fraction of incident radiant energy that is transferred to kinetic energy of charged
particles by interactions when traversing a distance dl in the material of density . If incident uncharged
particles of a given type and energy can produce several types of interactions in a material, tr/ can be
expressed in terms of the partial cross Sections, J, by the relation
tr N A
=

(4.3.5)

f J J
J

where fJ is the average fraction of the incident particle energy that is transferred to kinetic energy of
charged particles in an interaction of type J.
The mass energy transfer coefficient is related to the mass attenuation coefficient by
tr

= f =

f J J
J
J

(4.3.6)

For tr/ of a compound material the material is usually treated as consisting of independent atoms and
the contributions from the different components are summed considering their partial density.
A small part of the energy transferred to charged particles may not be locally absorbed in the material
but further transferred to secondary photons (e.g. Bremsstrahlung). Therefore, an additional coefficient,
the mass energy absorption coefficient, en/, is defined by the product of tr/ and (1-g) where g is the
fraction of the energy of charged particles that is lost in radiative processes in the material.
Data of mass energy transfer and mass energy absorption coefficients are given by Seltzer [93Se].
For neutron radiation, the kerma coefficient K/ (kerma per unit neutron fluence, often called kerma
factor) is mostly used instead of tr/ for characterising the energy transfer (see 4.4.1). Data of kerma
coefficients for biological important materials from thermal to 150 MeV neutrons are published by
Chadwick et al. [99Ch] and in ICRU Report 63 [00I1].

4.3.3 Mass stopping power and linear energy transfer (LET)


Charged particles passing matter loose energy by collisions with electrons, by emission of bremsstrahlung
in the electric fields of nuclei or atomic electrons or by elastic Coulomb scattering and inelastic nuclear
processes on atoms or nuclei. This effect is characterized by the mass stopping power S/ for charged
particles in a material with density . It is
S

1 dE
dl

unit: J m2 kg1

(4.3.7)
Landolt-Brnstein
New Series VIII/4

Ref. p. 4-27]

4 Radiological quantities and units

4-7

where dE is the energy lost by a charged particle in traversing a distance dl in the material. S =dE/dl is
called the linear stopping power. E may be given in eV and the unit of S/ may then expressed in
eV m2 kg1 or other multiples like MeV cm2 g1, for example.
The transfer of energy from the primary charged particle to secondary electrons is of specific interest
in dosimetry, especially to those electrons receiving a kinetic energy less than a given value only. They
will locally deposit their energy near to the track of the primary particle. This led to the definition of the
quantity linear energy transfer (LET) or restricted linear electronic stopping power L given by
L =

dE
dl

unit: J m1, often used keV m1

(4.3.8)

where dE is the energy lost by a charged particle due to electronic collisions when traversing a distance
dl minus the sum of the kinetic energies of all electrons released with kinetic energies in excess of . This
definition given in ICRU Report 60 [98I1] differs from earlier ones [80I1] in a way that L now includes
the binding energies for all collisions and the threshold of the kinetic energy of the released electrons is
now instead of minus the binding energy.
is often given in eV and then the notation L100 means an energy cutoff of 100 eV. L is often called
unrestricted linear energy transfer L and is equal to Sel, the electronic stopping power due to collisions
with electrons.

4.3.4 Mean energy expended in a gas per ion pair formed


In dosimetry, where often charge measurements due to ionisation in gases are the basis of dose
determinations, the kinetic particle energy necessary to create an ion pair is of general interest. This led to
the definition of the mean energy expended in a gas per ion pair formed W. It is
W=

E
N

unit: J

(4.3.9)

where N is the number of ion pairs when the initial kinetic energy E of the charged particle is completely
dissipated in the gas considered. This definition includes also the ions produced by secondary electrons or
bremsstrahlung.

4.4 Quantities related to energy transfer


4.4.1 Stochastic quantities
The energy transfer from incident particles to a target material is a stochastic process. For example, the
energy deposition along a track of a charged particle is randomly distributed. The values of stochastic
quantities are, therefore, subject to inherent fluctuations. They generally follow a probability distribution
and mean values may be given. For example, a Poisson distribution is already uniquely determined by its
mean value. Stochastic quantities are often used in microdosimetry in order to describe the energy
transfer to very small volumes.

Landolt-Brnstein
New Series VIII/4

4-8

4 Radiological quantities and units

[Ref. p. 4-27

4.4.1.1 Energy deposit and energy imparted


The energy deposit i, is the energy deposited in a single interaction i, thus

i = in out + Q

unit: J, often used eV

where in is the energy of the incident ionising particle (excluding rest energy), out the sum of energies of
all ionising particles leaving the interaction (excluding rest energy) and Q the change in the rest energies
of the nucleus and all particles involved in the interaction. Q > 0 means a decrease of rest energy, Q < 0
an increase.
The total energy transferred to matter in a given volume is often of interest. The energy imparted to
the matter in a given volume is the sum of all energy deposits in the volume

unit: J, often used eV

The mean energy imparted to the matter in a given volume is a non-stochastic quantity and can be
expressed in terms of the radiant energy Rin (sum of all radiant energies of the incoming particles) and Rout
(sum of the radiant energies of all outgoing particles). It is

= Rin Rout +

Q ,

unit: J, often used eV

4.4.1.2 Lineal energy and specific energy


Corresponding to the non-stochastic quantity LET the stochastic quantity lineal energy y is defined by the
quotient of s by l , where s is the energy imparted to the matter in a given volume by a single energy
deposition event and l is the mean chord length of that volume, thus

y=

s
l

unit: J m1, mostly used keV m1

This quantity is mainly used in microdosimetry, especially in measurements with low-pressure tissueequivalent proportional counters where single event distributions in terms of y are measured.
The specific energy (imparted) z is the quotient of by m, where is the energy imparted to matter of
mass m. It is

z=

unit: gray (Gy), 1 Gy = 1 J kg1

The specific energy includes the energy transferred to the matter m from all events involved.

4.4.2 Non-stochastic quantities


4.4.2.1 Kerma, kerma rate
The transfer of energy from uncharged particles (indirectly ionising particles, e.g. photons or neutrons) to
matter is performed by the liberation and slowing down of secondary charged particles in this matter. This
led to the definition of the quantity kerma. The kerma K is the quotient of dEtr by dm, where dEtr is the
sum of the kinetic energies of all charged particles liberated by uncharged particles in a mass dm of
material. It is given by
K=

dE tr ,
dm

unit: gray (Gy), 1 Gy = 1 J kg-1

Landolt-Brnstein
New Series VIII/4

Ref. p. 4-27]

4 Radiological quantities and units

4-9

Kerma is a non-stochastic quantity. For a very small mass element, however, the energy transfer dEtr
underlies in principle stochastic fluctuations. In this case a non-stochastic quantity means that dEtr is seen
to be the expectation value of the sum of energies of liberated charged particles.
For monoenergetic uncharged particles of energy E the kerma is related to the fluence by
K = E (tr/)

(4.4.1)

For a given energy distribution E of the uncharged particles the kerma can be calculated by
K = E E (tr/) dE

(4.4.2)

For neutrons, the quotient of K by , is called kerma coefficient (often also called kerma factor) where
is the neutron fluence (see 4.3.2).
The kerma rate, K& , is the quotient of dK by dt, where dK is the increment of K in the time interval dt.
dK ,
K& =
dt

unit: Gy s1.

4.4.2.2 Absorbed dose, absorbed dose rate


The quantity absorbed dose is a basic quantity in radiation dosimetry and relevant to all types of ionising
radiation whether directly or indirectly ionising. It is a non-stochastic quantity and defined by:
D=

unit: gray (Gy), 1 Gy = 1 J kg1

dm

where is the mean energy imparted to the matter of mass dm.


While kerma is related only to those secondary charged particles produced in dm but transferring their
energy to matter partially also outside dm, absorbed dose includes all energy transferred to dm partially
also from secondary charged particles produced outside but entering dm. Only under charged particle
equilibrium and negligible radiation losses, however, the values of absorbed dose and kerma are equal in
a homogeneous material.
The absorbed dose rate, D& , is the quotient of dD by dt, where dD is the increment of the absorbed
dose in the time interval dt. It is
dD
D& =
dt

unit: Gy s1

4.4.2.3 Exposure, exposure rate


The quantity exposure is related to the production of charges in gas by ionising radiation. Historically its
definition is elder than kerma or absorbed dose. Its use, however, is restricted to photons only. The
exposure X is the quotient of dQ by dm, where dQ is the absolute value of the total charge of the ions of
one sign produced in air when all the electrons and positrons liberated or created by photons in air of
mass dm are stopped in air.

X =

dQ
dm

unit: C kg1 (former: roentgen, R)

It should be noted that in this definition the charges due to ionisation arising from the absorption of
bremsstrahlung emitted by the electrons is not included in dQ.
Landolt-Brnstein
New Series VIII/4

4-10

4 Radiological quantities and units

[Ref. p. 4-27

The exposure rate, X& , is the quotient of dX by dt, where dX is the increment of the exposure in the
time interval dt. It is

dX
X& =
dt

unit: C kg1 s1

4.5 Dose quantities in radiation protection


4.5.1 Concept of radiation protection quantities
The development of dosimetric concepts and the definition of specific quantities for use in radiation
protection have a long history. An important basis for the present concepts was already provided in the
60's and 70's by both the International Commission on Radiological Protection (ICRP) and the
International Commission on Radiation Units and Measurements (ICRU). In 1991 in its Publication 60
[91I1], the ICRP has published its most recent general recommendations for radiation protection
including a system of quantities.
The ICRP and ICRU have developed a hierarchy of quantities for radiation protection applications
comprising primary limiting dose quantities (called protection quantities) taking account of human
body properties and operational dose quantities for monitoring of external exposure. For monitoring of
internal exposure other quantities than dose quantities are used.
The basic idea of a primary limiting quantity is to relate the risk of exposure to ionising radiation
(exposure by internal and external radiation sources) to a single (dose) quantity which takes account of
the man as a receptor, the different radiation sensitivities of various organs and tissues and the different
radiation qualities. Other influence parameters, however, e.g. the influence of dose and dose rate or sex
and age of a person exposed on the biological response and the exposure risk, were not explicitly
considered in the definition of these quantities.
Operational quantities are dose equivalent quantities defined for use in radiation protection
measurements related to external exposure (area or individual monitoring). They are needed for
monitoring external exposures because

protection quantities are generally not measurable,


for area monitoring a point quantity is needed,
a non-isotropic human-body related quantity like effective dose is not appropriate in area monitoring,
instruments for radiation monitoring need to be calibrated in terms of an operational quantity.

Operational quantities usually provide an estimate or upper limit for the value of the limiting quantities
due to an exposed, or potentially exposed, person under most irradiation conditions. They are often used
in practical regulations instead of the primary limiting quantities.
For internal exposure, however, other methods are used and no similar dose quantities have been
defined. In this case organ doses or effective dose are estimated from the information on intake or
excretion of radioactive substances. Model based conversion coefficients exist for a large number of
radionuclides relating the intake to organ doses and effective dose (see 4.6 and Chapter 7).
Both, protection and operational quantities can be related to radiation field quantities (see Sect. 4.2)
or air kerma (see Sect. 4.3) which are point quantities defined in any point of a radiation field and whose
units are directly realised through primary standards at national standards laboratories since long time.
The numerical relations (conversion coefficients) between those quantities and the protection or
operational dose quantities are given in Chapter 6.

Landolt-Brnstein
New Series VIII/4

Ref. p. 4-27]

4 Radiological quantities and units

4-11

4.5.2 Protection quantities


In 1977 the ICRP [77I1] introduced the tissue (or organ) dose equivalent HT and the effective dose
equivalent HE whose definition takes care of the relative variation of the tissue response with different
types of radiation and different tissues or organs in the human body by introducing tissue weighting
factors [73Jac]. Although in general this concept was not changed by ICRP 60 [91I1] in 1990, important
modifications, however, were introduced e.g. replacing dose equivalent quantities by equivalent dose
quantities. The present system of quantities is summarised in the following.
4.5.2.1 Absorbed dose and equivalent dose in a tissue or organ
The absorbed dose in a tissue or organ DT is the absorbed dose averaged over the volume of a tissue or
organ T (rather than at a point). While the absorbed dose at a point generally is the fundamental dose
quantity, in radiation protection the mean dose in an organ becomes the basic protection quantity
correlated with the exposure risk. This concept is obviously based on the linear dose-effect relationship
and the additivity of doses for risk assessment as an appropriate approximation in the low dose range.
The equivalent dose in a tissue or organ is defined by
HT =

w D
R

T, R

unit: sievert (Sv) (1 Sv = 1 J kg1)

where DT,R is the mean organ dose in the tissue or organ T from radiation of type R incident on the human
body and wR are radiation weighting factors characterising the biological effectiveness of the specific
radiation R relative to photons. These factors have replaced the mean quality factors used in the concept
of organ dose equivalent before [77I1]. The sum is taken over all types of radiation involved.
4.5.2.2 Radiation weighting factors
For external irradiation, the values of the radiation weighting factors wR are given by the parameters of
the external radiation field only (type and energy distribution of the radiation incident on the body). This
means that wR is a body-averaged value representing a mean value for the relative biological effectiveness
of all tissues of the body and any local variation of the radiation quality in the human body which may
result from the generation of secondary radiation of different types in the body, is not explicitely
considered. This effect is mainly important in the case of incident neutrons where at low energies
secondary photons strongly contribute to the absorbed doses of various organs.
The wR values for various types of radiation are specified in ICRP 60 in a table (see Table 4.2). For
photons, electrons and muons of all energies a value of one is fixed with the exception of Auger electrons
emitted from nuclei bound to DNA. For this case there exists no ICRP recommendation until now.
The radiation weighting factor for neutrons depends on the neutron energy. Different wR values are
given by either a step function or a continuous function as an approximation (see Fig. 4.1). In practice,
neutron fields contain neutrons with a broad energy distribution. Because the use of a continuous wRfunction for effective dose estimation is more appropriate in these cases it is recommended to apply the
continuous function in any case to avoid ambiguities. Then the weighting factor for neutrons ranges from
5 to 22 depending on neutron energy with its maximum value at 500 keV. All conversion coefficients for
neutrons published in ICRP 74 [96I1] and ICRU 54 [98I2] are based on the continuous function only (see
Chapt. 6).

Landolt-Brnstein
New Series VIII/4

4-12

4 Radiological quantities and units

[Ref. p. 4-27

Table 4.2. Radiation weighting factors wR


Radiation

wR

Photons
Electrons1), muons

1
1

Neutrons:
En
En
En
En
En

5
10
20
10
5

<10 keV
=10 keV to 100 keV
>100 keV to 2 MeV
>2 MeV to 20 MeV
>20 MeV

Protons: Ep

> 2 MeV
(unless recoil protons)

-particles, fission fragments, heavy nuclei

20

As an approximation to the step function


introduced for neutrons ICRP has
specified a smooth wR function:
wR = 5 + 17 exp ([ln(2 En)]2/6)
with En neutron energy in MeV.

1) With the exception of Auger electrons from atoms bound to DNA

The radiation weighting factor for incident external protons with energies above 2 MeV has been set
to 5. It is, however, questioned if this value is appropriate for protons of all energies above 2 MeV. There
exists a general opinion that a weighting factor of about 2 seems to be more realistic for high energy
protons above about 5 to 10 MeV. External protons of lower energies have a small range in tissue and
contribute to the skin dose only.
4.5.2.3 Effective dose
The effective dose E is the weighted sum of the equivalent doses in tissues and organs T:
E=

w H
T

with

=1

unit: sievert (Sv)

(4.5.1)

where wT are tissue weighting factors characterising the relative sensitivity of the various tissues with
respect to stochastic effects resulting from ionising radiation exposure and HT is the equivalent dose in
one of the 13 specified tissues and organs (see Sect. 4.5.2.4).
The effective dose is a quantity which is not sex specific or dependent on age of a person. In principle,
the effective dose is determined by taking the dose values in all tissues and organs of an individual
person. Those data, however, are never measurable. For external exposure, therefore, always calculated
conversion coefficients are used which relate the external radiation field to the doses in the tissues and
organs (see Chapt. 6).
Following ICRP Report 74 [96I1], the effective dose is then calculated by
E = wbreast H breast, female +

T breast

wT

H T, male + H T, female
2

(4.5.2)

Under a given exposure condition (radiation field, direction of radiation incidence, exposure period),
therefore, all persons are given the same effective dose value independent of sex and age.
Landolt-Brnstein
New Series VIII/4

Ref. p. 4-27]

4 Radiological quantities and units

4-13

4.5.2.4 Tissue Weighting Factors


The definition of effective dose takes care of the different radiosensitivity of the various organs and
tissues in the human body with respect to cancer induction and mortality by introducing tissue weighting
factors. Twelve tissues and organs are specified with individual weighting factors wT. The values have
been developed from a reference population of equal numbers of both sexes and a wide range of ages.
They are applied to workers, to the whole population, and to either sex including children and the unborn
child (foetus).
An additional remainder tissue with a weighting factor of 0.05 is also defined [91I1]. Its dose is
given by the mean value from ten specified tissues and organs (see Table 4.3). The upper large intestine
formerly included in the remainder, is now considered as part of the colon and has been replaced by the
extrathoracic airways [93I3, 94I2]. While in the calculation of conversion coefficients for the intake of
radionuclides the remainder dose is obtained from the mass-weighted doses to the single tissues and
organs involved, the coefficients for external exposure are calculated giving identical weights to each of
the remainder tissues [96I1].
Table 4.3. Tissue weighting factors wT
Organ or tissue
Gonads
Bone marrow (red)
Colon
Lung
Stomach
Bladder
Breast
Liver
Oesophagus
Thyroid
Skin
Bone surface
Remainder1)

wT
0.20
0.12
0.12
0.12
0.12
0.05
0.05
0.05
0.05
0.05
0.01
0.01
0.05

1) Remainder tissues are adrenals, brain, extrathoracic


airways, small intestine, kidney, muscle, pancreas,
spleen, thymus and uterus.
The mean value of the equivalent doses of the ten
remainder organs and tissues is to be multiplied by
0.05. If in a special case a single tissue or organ has an
equivalent dose higher than each of the 12 individually
defined organs and tissues, then this organ or tissue
should get a weighting factor of 0.025 and the other 9
remainder tissues together a weighting factor of 0.025.

4.5.2.5 Committed or collective equivalent dose and effective dose


Several subsidiary dosimetric quantities have been additionally defined. After an intake of radionuclides
to a body these nuclides may give rise to equivalent doses in different tissues and organs of the body
spread over long time depending on the physical and biological half-life of the radionuclides and their
biokinetic behaviour in the body.
The time integral of the equivalent dose rate is called the committed equivalent dose HT( ), in a tissue
or organ T, where is the integration time (in years) following the intake at time t0.
t0 +

H T ( ) =

H&

( t )dt

(4.5.3)

t0

If is not specified, it is implied that its value is 50 y for workers and from intake up to age 70 years for
members of the public including children. For patients in nuclear medicine, the integration may run from
t0 to because the biological and physical half-life of the radionuclides applied is much less than 10 y.

Landolt-Brnstein
New Series VIII/4

4-14

4 Radiological quantities and units

[Ref. p. 4-27

The same specification holds for the quantity committed effective dose E( ) defined by the weighted
sum of HT( ) over all specified tissues and organs T.
E( ) =

H T ( )

(4.5.4)

All dosimetric quantities referred before are related to a single tissue or organ of a single individual.
Often it may be of interest to quantify the total dose a number of people received from one source or one
release of radioactive material. The relevant quantity is called collective equivalent dose ST in a tissue or
organ T and is defined by

ST = H T
0

dN
dH T
dH T

unit: man sievert (man Sv)

(4.5.5)

where (dN/dHT)dHT is the number of individuals receiving an equivalent dose between HT and HT+dHT or
by
ST =

T, i

(4.5.6)

Ni

where Ni is the number of individuals in a subgroup i receiving a mean tissue equivalent dose HT,i . The
summed effective doses of all members of a group or population is called the collective effective dose S
defined in a similar way by

S = E
0

dN
dE
dE

or

S=

E N
i

unit: man Sv

(4.5.7a+b)

where Ni is a subgroup i receiving a mean equivalent dose Ei .

4.5.3 Operational quantities


4.5.3.1 Dose equivalent and quality factor
The radiation protection quantity dose equivalent H is defined by
unit: Sv (1 Sv = 1 J kg1)

H=QD

where D is the absorbed dose at the point of interest and Q a quality factor weighting the relative
biological effectiveness of radiation. Q is defined as a function of the linear energy transfer L of a charged
particle in water [77I1]. In principle, this concept of Q has not been changed by ICRP 60 [91I1], but the
dose equivalent is now restricted to the definition of operational radiation protection quantities and the
quality factor function Q(L) was modified in 1991 according to the following equation:

Q(L) =

1
0.32 L 2.2
300/L

for
L < 10 keV/m
for 10 L 100 keV/m
for
L > 100 keV/m

(4.5.8)

Landolt-Brnstein
New Series VIII/4

Ref. p. 4-27]

4 Radiological quantities and units

4-15

The quality factor Q at a point in tissue is then given by [86I1]:

Q=

1
Q( L )DL dL
D L=0

(4.5.9)

where DL is the distribution of D in L at the point of interest. This function is most important for neutrons
because various types of secondary charged particles are produced in tissue in this case.
4.5.3.2 The concept of operational quantities
The basic concept of the operational quantities is described in the ICRU Reports 39 and 43 [85I1, 88I1].
They have been introduced linking the external irradiation to the effective dose and the equivalent dose of
the skin and eye lens in order to control their limits. The present definitions are given in ICRU Report 51
[93I1]. The operational quantities for radiation protection are dose equivalent quantities defined either for
strongly penetrating or for weakly penetrating radiation incident on the human body (sometimes also the
expressions penetrating and low penetrating are used instead of strongly and weakly penetrating
radiation).
The radiation is characterised as either weakly- or strongly penetrating depending on which dose
(effective dose or skin equivalent dose) is closer to the corresponding limit. Weakly penetrating radiations
are -particles, -particles with energies below 2 MeV and photons with mean energies below about 12
keV. Photons above this energy, electrons above 2 MeV and all neutrons are strongly penetrating
radiation.
Due to the different tasks in radiation protection monitoring area monitoring for controlling the
radiation at work places and definition of controlled or forbidden areas or individual monitoring for the
control and limitation of individual exposures different operational quantities were defined. While
measurements with an area monitor are mostly performed free in air, an individual dosemeter is usually
worn on the front of the body. As a consequence, in a given situation, the radiation field seen by an area
monitor free in air differs from that seen by an individual dosemeter worn on a body where the
radiation field is strongly influenced by the backscatter and absorption of radiation in the body. The
operational quantities allows for this effect. They may be presented as follows:

Radiation type

Operational quantities for


area monitoring
individual monitoring

Strongly penetrating radiation


Weakly penetrating radiation

ambient dose equivalent


directional dose equivalent

personal dose equivalent


personal dose equivalent

4.5.3.3 Operational quantities for area monitoring


ICRU sphere phantom
For all types of radiation the operational quantities for area monitoring are defined on the basis of a dose
equivalent value at a point in a simple phantom, the ICRU sphere. It is a sphere of tissue-equivalent
material (30 cm in diameter, density: 1 g cm3, mass composition: 76.2 % oxygen, 11.1 % carbon, 10.1 %
hydrogen and 2.6 % nitrogen). It adequately approximates the human body as regards the scattering and
attenuation of the radiation fields under consideration.

Landolt-Brnstein
New Series VIII/4

4-16

4 Radiological quantities and units

[Ref. p. 4-27

Aligned and expanded radiation field


The operational quantities for area monitoring defined in the ICRU sphere should retain their character of
a point quantity and the property of additivity. This is achieved by introducing the terms expanded and
aligned radiation field in the definition of these quantities (see Fig. 4.1).


















UHDOILHOG






DOLJQHGILHOG

 























DOLJQHGDQGH[SDQGHGILHOG

Fig. 4.1. Aligned and expanded field concept.

An expanded radiation field is a radiation field in which the spectral and the angular fluence have the
same values in all points of a sufficiently large volume equal to the values in the actual field at the point
of interest. The expansion of the radiation field ensures that the whole ICRU sphere is thought to be
exposed to a homogeneous radiation field with the same fluence, energy distribution and directional
distribution as in the point of interest of the real radiation field.
If all radiation is (thought to be) aligned in the expanded radiation field so that it is opposed to a radius
vector specified for the ICRU sphere, the aligned and expanded radiation field is obtained. In this
fictitious radiation field, the ICRU sphere is homogeneously irradiated from one direction, and the
fluence of the field is the integral of the angular differential fluence at the point of interest in the real
radiation field over all directions. In the expanded and aligned radiation field, the value of the dose
equivalent at any point in the ICRU sphere is independent of the directional distribution of the radiation
of the real radiation field.
Ambient dose equivalent H*(d )
For area monitoring of strongly penetrating radiation the operational quantity is the ambient dose
equivalent H*(10) defined by:

Landolt-Brnstein
New Series VIII/4

Ref. p. 4-27]

4 Radiological quantities and units

4-17

The ambient dose equivalent H*(d) at a point of interest in the real radiation field, is the dose
equivalent that would be produced by the corresponding aligned and expanded radiation field, in
the ICRU sphere at a depth d, on the radius vector opposing the direction of radiation incidence.
For strongly penetrating radiation it is d = 10 mm and H*(d) is written H*(10).
While this definition with the parameter d is given in ICRU [93I1] and ICRP [91I1] the most recent
ICRU Report [01I1] dealing also with operational quantities defines ambient dose equivalent by
H*(10), thus restricting its definition to strongly penetrating radiation only. In practice, however, this has
already been realised because other values have never been used.
As a result of the imaginary alignment and expansion of the radiation field, the contributions of
radiation from all directions add up. The value of H*(10) is therefore independent of the directional
distribution of the radiation in the actual field. This means that the reading of an area dosemeter for the
measurement of H*(10) should be independent of the directional distribution of the radiation an ideal
detector should have an isotropic fluence response.
Directional dose equivalent, H'(d , )
For area monitoring of weakly penetrating radiation the operational quantity is the directional dose
equivalent H'(0.07, ) or, in rare cases, H'(3, ) defined by.
The directional dose equivalent H'(d, ) at a point of interest in the actual radiation field, is the
dose equivalent that would be produced by the corresponding expanded radiation field, in the
ICRU sphere at a depth d, on a radius in a specified direction .
For weakly penetrating radiation it is d = 0.07 mm and H'(d, ) is written H'(0.07, ).
In case of monitoring the dose to the eye lens H'(3, ) with d = 3 mm may be chosen.
In practice H'(0.07, ) is almost exclusively used in area monitoring for weakly penetrating radiation.
For unidirectional radiation incidence the quantity may be written H'(0.07,), where is the angle
between the direction and the direction opposite to radiation incidence.
The value of the directional dose equivalent can strongly depend on the direction . The same is true
for instruments for measuring weakly penetrating radiation e.g. beta- or alpha-particle radiation the
reading of which can strongly depend on the orientation in space. In radiation protection practice,
however, it is always the maximum value of H'(0.07, ) at the point of interest which is of importance. It
is usually obtained by rotating the dose rate meter during the measurement and looking for the maximum
reading.
4.5.3.4 Operational quantities for individual monitoring
Individual monitoring is usually performed with individual dosemeters worn on the body and the
operational quantity defined for this application takes into account this situation. The true value of the
operational quantity is determined by the irradiation situation near the point where the dosemeter is worn.
For individual monitoring the operational quantity is the personal dose equivalent Hp(d).
The personal dose equivalent Hp(d) is the dose equivalent in ICRU tissue at a depth d in a human
body below the position where an individual dosemeter is worn.
For strongly penetrating radiation a depth d = 10 mm is recommended.
For weakly penetrating radiation a depth d = 0.07 mm is recommended.
In special cases of monitoring the dose to the eye lens a depth d = 3 mm may be appropriate.
Landolt-Brnstein
New Series VIII/4

4-18

4 Radiological quantities and units

[Ref. p. 4-27

The operational quantities for individual monitoring meet several criteria. They are defined for all
types of radiation, additive with respect to various directions of radiation incidence, take into account the
backscattering from the body and can be measured with a dosemeter worn on the body. The personal dose
equivalent quantities, Hp(10) and Hp(0.07), are defined in the person, in the actually existing radiation
field, and are measured directly on the person.
Other requirements the quantities should satisfy can, however, be fulfilled only with additional
specifications. An operational quantity for individual monitoring should allow the effective dose to be
assessed or should provide a conservative estimate under nearly all irradiation conditions. This, however,
requires that the personal dosemeter must be worn at a position on the body which is representative with
respect to the exposure. For the usual dosemeter position in front of the trunk the quantity Hp(10) mostly
furnishes a conservative estimate of E even in cases of lateral or isotropic radiation incidence on the body.
In cases of exposure from the back, however, a dosemeter worn at the front side and correctly measuring
Hp(10), will not provide a conservative estimate of E.
A further requirement for an operational quantity is that it allows dosemeters to be calibrated under
reference conditions in terms of that quantity. The personal dose equivalent is defined in the individual
human body and obviously individual dosemeters cannot be calibrated in front of a real human body. For
calibration, the human body must therefore be replaced by an appropriate phantom. Three standard
phantoms have been defined by ISO for this purpose and the definition of Hp(10) and Hp(0.07) is
extended to be defined not only in the human body but also in three phantoms of ICRU tissue (see
Fig. 4.2) a slab phantom (30 cm 30 cm 15 cm), a wrist phantom (a cylinder of 73 mm in diameter
and 300 mm in length) and a finger phantom (a cylinder of 19 mm in diameter and 300 mm in length). In
reference radiation fields used for calibration, the values of the quantities in these phantoms, Hp,slab(10)
and Hp,slab(0.07) etc., are defined as the true values of the corresponding Hp-quantities (see also Sect. 6.1.2
and Sect. 10.2.1).

Fig. 4.2. Phantoms of ICRU tissue for the


definition of Hp-quantities for calibration of
individual dosemeters.
a) slab phantom
b) wrist phantom
c) finger phantom

4.6 Radioactivity quantities


The decay of a radionuclide is a stochastic process which means that the number of decays within a fixed
time interval is described by a probability distribution. The expectation value of the number of decays is
determined by the decay constant which is specific for each radionuclide and energy state (mostly the
decay constant for the ground state is given).
The decay constant of a radionuclide in a particular energy state is the quotient of dP by dt, where
dP is the probability that a given nucleus undergoes a spontaneous transition from that energy state in the
time interval dt. It is

dP
dt

unit: s1
Landolt-Brnstein
New Series VIII/4

Ref. p. 4-27]

4 Radiological quantities and units

4-19

The half-life T1/2 of a radionuclide in a particular energy state is the mean time of the radionuclide in
that state to decrease to one half of their initial number of nuclei. It is T1/2 = (ln 2)/.

4.6.1 Activity, specific activity, activity concentration, activity per area


The activity A of an amount of a radionuclide in a particular energy state at a given time is the quotient of
dN by dt, where dN is the expectation value of the number of spontaneous nuclear transitions from that
energy state in the time interval dt. It is
A=

dN
dt

unit: becquerel (Bq), 1 Bq = 1 s1

Radionuclides are mostly included in other solid, liquid or gaseous material and the amount is
quantified by the quantities specific activity and activity concentration.
The specific activity as is given by the quotient of the activity A by the mass m, where A is the activity
of the radionuclide in the mass m.
as =

A
m

unit: Bq kg1

The activity concentration cnuclide is given by the quotient of the activity A by the volume V, where A is
the activity of the radionuclide in the volume V.
c nuclide =

A,
V

unit: Bq m3

For the determination of contaminations the distribution of radionuclides on surfaces is of interest.


The related quantity is the activity per unit area aa defined by the quotient of the activity A by the area F,
where A is the activity of a radionuclide distributed on the surface area F.
aa =

A
F

unit: Bq m2, often Bq cm2

For decontamination of a surface from deposited radionuclides it is usually important if the


radionuclides are removable or if they are diffused into the surface region of the material and are fixed
near the surface in the material. If an aa-value is given it should be specified if this value is related to the
removable part only or to the total activity at the surface.

4.6.2 Specific quantities for radon, thoron and their progeny


Radon (222Rn) and thoron (220Rn) are gaseous radionuclides in the U- and Th-decay chain, respectively,
occurring naturally (see 3.4.3). Their decay products are also radionuclides but metallic. While for radon
the short-lived progeny 218Po, 214Pb, 214Bi and 214Po (see Table 4.4) are important in radiation protection,
the important thoron progeny are 216Po, 212Pb, 212Bi and 212Po (see Table 4.5). In air there is usually a
mixture of radon/thoron and short-lived radon/thoron progeny. These progeny are mostly attached to
aerosols. A few percentages of them, however, are non-attached. The progeny may be deposited in the
lung where its decay by alpha-particle emission is seen to be most important for lung cancer induction.
Specific quantities have been defined taking care of this situation.
Landolt-Brnstein
New Series VIII/4

4-20

4 Radiological quantities and units

[Ref. p. 4-27

Table 4.4. Data of radon (222Ra) progeny (nuclear data are from [NN98])
potential alpha energy
number of
Radionuclide half-life
atoms per Bq
per atom
per Bq /
A/
i
[MeV] [1012 J] [MeV]
T1/2
[1012 J]
218

Po
Pb
214
Bi
214
Po
214

1)
2)

1
2
3
4

3.10 min
26.8 min
19.9 min

268
2 320
1 723

164 s

1)

13.69
7.69
7.69
7.69

2.19
1.23
1.23
1.23

3 670
17 800
13 100

589
2 860
2 100

0.106
0.513
0.381

2103

2.9104

6108

no number is given because all atoms decay in less than 1 s and a calculated number would be much less than 1.
factor k is defined in Eq. (4.6.1)

Table 4.5. Data of thoron (220Ra) progeny (nuclear data are from [NN98])
potential alpha energy
number of
Radionuclide half-life
atoms per Bq
per atom
per Bq /
A/
i
[MeV]
[1012 J]
[MeV] [1012 J]
T1/2
216

Po

0.15 s

1)

Pb
Bi
212
Po

2
3
4

10.6 h
60.6 min
304 ns

55 056
5 246

212

212

1)
2)
3)

k(2)

1)

14.6
7.8
7.82)
8.8

2.34

3.32

0.51

1.25
1.25
1.25

429 000
40 900

68 710
6 554

3.8106

6107

k(3)

6106
0.913
0.087
81012

no number is given because all atoms decay in less than 1 s and a calculated number would be much less than 1.
mean value from decay of 212Bi and 212Po by -particle emission.
factor k is defined in Eq. (4.6.1)

Potential alpha energy


The potential alpha energy i of an atom i in the decay chain of radon or thoron is the total energy of
alpha-particles emitted during the decay of this atom to the long-living 210Pb or stable 208Pb, respectively.
The potential alpha energy of N atoms of type i is Ni. The number of atoms N per Bq is equal to A/,
where A is the activity of this radionuclide and its decay constant. The potential alpha energy per Bq is
then given by / (unit: J Bq1, often used MeV Bq1).
Concentration in air
The potential alpha energy concentration cp,i of a short-lived radon (or thoron) progeny in air is the sum
of the potential alpha energy i of all atoms of this progeny present in a volume V divided by this volume.
It is
cp,i =

N i i

= ci i
V
i

unit: J m3 often MeV m3

where Ni is the number of atoms of this progeny in the volume V, ci the corresponding activity
concentration and i the decay constant. The units are related by 1 J m3 = 6.242 1012 MeV m3.
The potential alpha energy concentration (PAEC) cp of any mixture of short-lived radon (or thoron)
progeny in air is the sum of the potential alpha energy concentrations of all progeny in the volume
considered.
Landolt-Brnstein
New Series VIII/4

Ref. p. 4-27]
cp =

4 Radiological quantities and units

c = c
p,i

4-21

unit: J m3 often MeV m3

Historically, for the potential alpha energy concentration the unit working level (WL) has widely been
used. While originally defined as the potential alpha energy concentration associated with the radon
progeny in equilibrium with 100 pCi l1, 1 WL is now accurately fixed equal to 1.300 108 MeV m3
which equals 2.08 105 J m3.
Equilibrium equivalent concentration, equilibrium factor
In case of radioactive equilibrium the activity concentration of radon cRn and of its progeny are equal.
This, however, is usually not the case in air. For a non-equilibrium mixture a quantity equilibrium
equivalent concentration ce has been defined.
The equilibrium equivalent concentration (EEC) ce corresponding to a non-equilibrium mixture of
progeny in air is the fictitious activity concentration of radon in radioactive equilibrium with its shortlived progeny that has the same potential alpha energy concentration cp as the actual non-equilibrium
mixture. It is always ce cRn. The SI-unit for both quantities, ce and cRn, is Bq m3. In order to avoid
confusion, the values of ce are often marked Bq m3 (EEC).
The equilibrium equivalent concentration ce can be calculated from the activity concentrations of the
progeny by the equation
ce =

k c
i

with ki = ( i i )

i )

(4.6.1)

The factors ki are given in Tables 4 and 5 and it is


for radon progeny:
and for thoron progenies:

ce = 0.106 cPo-218 + 0.513 cPb-214 + 0.381 cBi-214 + 6 108 cPo-214


6

ce = 7 10 cPo-216 + 0.913 cPb-212 + 0.087 cBi-212 + 8 10

12

cPo-212

(4.6.2)
(4.6.3)

Obviously, the radionuclides 216Po, 214Po and 212Po can be ignored when calculating ce because of their
very low kivalues.
The equilibrium factor F is defined as the quotient of the equilibrium equivalent concentration and the
activity concentration of the parent nuclide, radon, in air.
F = ce / cRn

(4.6.4)

The value of F ranges from 0 to 1 and is a measure to what extent radioactive equilibrium between radon
and its progeny is obtained. Mostly this is not the case and often a mean value of 0.4 is convenient for the
situation in homes.
The unattached progeny in air which are not attached to aerosols is also of special interest. The
unattached fraction fp is defined by the relative fraction of the total potential alpha energy concentration
which stems from progeny in air which are not attached to aerosols. It is
fp =

cpf
cp

cpf
cpa + cpf

(4.6.5)

where c pa is the potential alpha energy concentration of the progeny attached to aerosols, c pf is that of the
unattached fraction and cp is the sum of both parts.
Landolt-Brnstein
New Series VIII/4

4-22

4 Radiological quantities and units

[Ref. p. 4-27

Inhalation exposure of individuals


The exposure of an individual to radon progeny Pp is defined as the time integral of the potential alpha
energy concentration cp in air to which the individual is exposed.
T

Pp (T ) = cp (t ) dt

(4.6.6)

where T is the period of the exposure. A similar integral is given if the equilibrium concentration ce(t) is
taken for integration. It is then called the equilibrium equivalent exposure Pe(T):
T

Pe (T ) = ce (t ) dt

(4.6.7)

The potential alpha energy exposure Pp is often expressed in terms of working level month (WLM),
even if not recommended for further use. This quantity has been introduced especially for specifying
occupational exposure and a fixed time period T of 170 hours has therefore been chosen equal to a mean
monthly working time. The relation to SI-units (see Table 4.1) is given by 1 WLM = 3.54 103 J h m-3 =
2.21 1010 MeV h m3.

4.7 Quantities for internal dosimetry


Internal exposure means an exposure by ionising radiation emitted from radionuclides incorporated and
distributed in the body. A direct measurement of doses in a human body is not possible. For internal
exposure there are, therefore, no specific operational dose quantities defined. In contrast to external
monitoring usually the committed tissue or organ equivalent doses and committed effective dose are
determined (period of 50 y for workers and a period up to the 70th year of life for members of the public
including children) and complex compartment models are used to describe the long term biokinetic
behaviour of the radionuclides in the human body (see Chapt. 7). The committed tissue and organ
equivalent doses of an individual are usually determined by external measurements, e.g. activity
concentration of specific radionuclides in the air, specific activity of food and water or contamination of
the skin, and the application of calculated dose conversion coefficients (often called dose coefficients)
which have been published for inhalation, ingestion and intake through the skin for a large number of
radionuclides (see Chapter 7). Measured excretion data are usually used to estimate the intake of
radionuclides subsequently and then conversion coefficients are applied to evaluate doses.
The intake of radionuclides by inhalation, ingestion or through intact or wounded skin or the excretion
by exhalation, urine, faeces etc, is determined in terms of measurable quantities. These are often activity
concentrations in air , cnuclide (in terms of Bq m-3), and inhalation frequencies or inhaled activities, the
specific activity of solids and liquids, as (in terms of Bq kg-1), the amount of ingested radioactive
substances or their specific activity in excretions. Further details are given in Chapter 7.
In cases where radionuclides emit high energy -rays their distribution in the body may be determined
by external measurements of -rays with a whole-body counter (large -detectors well shielded against
radiation from the environment) in combination with computer codes simulating the photon absorption in
the body.

Landolt-Brnstein
New Series VIII/4

Ref. p. 4-27]

4 Radiological quantities and units

4-23

4.8 Limits, constraints, action levels


The system of radiation protection as recommended by the ICRP [91I1] is based on the following
principles (see also Chapter 1):
a) No practice involving exposure to radiation should be adopted unless it produces sufficient benefit to
the exposed individuals or to society to offset the radiation detriment it causes (principle of
justification).
b) In relation to any particular source within a practice, the magnitude of individual doses, the number
of people exposed, and the likelihood of incurring exposures where these are not certain to be
received should all be kept as low as reasonably achievable, economic and social factors being taken
into account. This procedure should be constrained by restrictions on the doses to individuals (dose
constraints), or the risks to individuals in the case of potential exposures (risk constraints), so as to
limit the inequity likely to result from the inherent economic and social judgements (the optimisation
of protection).
c) The exposure of individuals resulting from the combination of all the relevant practices should be
subject to dose limits, or to some control of risk in the case of potential exposures. These are aimed at
ensuring that no individual is exposed to radiation risks that are judged to be unacceptable from these
practices in any normal circumstances. Not all sources are susceptible of control by action at the
source and it is necessary to specify the sources to be included as relevant before selecting a dose
limit (individual dose and risk limits).
Generally, radiation protection takes care of exposure situations and doses which are relevant for the
health of the persons involved or may not be ignored compared to the normal exposure from natural
radiation sources. This means that there should exist a dose level below which exposures from
radionuclides or other radiation sources may not be taken care of and where no regulations are necessary,
independent of the fact that, in principle, any radiation may induce cancer or genetic defects. The ICRP
sees such a dose range below a few tens of Sv (committed dose or dose per year) for a single individual
which is about 1/100 of the normal exposure from natural sources in the environment. Often an upper
boundary of 10 Sv (committed dose or dose per year) is used to decide if further investigations or
actions are necessary.
Usually the human exposures are classified in three different categories.
The first is called occupational exposure which means any exposure incurred at work and principally
as a result of situations which can be reasonably regarded as being in the responsibility of the operating
management. It also includes potential exposures where the probability of a future exposure due to
planned work forces may be estimated [97I1].
Medical exposures describe the exposure of patients during diagnostic and treatment. While medical
exposures are intended to provide a direct benefit to the patient, the practice should be justified and
optimized with respect to applied doses and the medical benefit.
Public exposures are all exposures other than occupational or medical exposures. Public exposures
include environmental exposures due to natural sources in the environment, e.g. natural actinides, radon,
potassium-40 and cosmic radiation, but also those exposures due to artificial sources where the target
group is the general population (details are given in Chapter 11). Examples are the broadly distributed
radionuclides from the nuclear bomb test in 1950 to 1970 and the contamination due to the Chernobyl
accident.

Landolt-Brnstein
New Series VIII/4

4-24

4 Radiological quantities and units

[Ref. p. 4-27

Dose limits
Dose limits have been recommended by the ICRP [91I1] for occupational exposure and for exposures to
the public. They are given in terms of effective dose and few organ equivalent doses, always summed
over a given period. The limits apply to the sum of the relevant doses from external exposure in the
specified period (often one year) and from intakes of radionuclides in the same period. The corresponding
internal dose is the 50-year committed dose (for occupational exposure) or the committed dose up to the
age of 70 years (for members of the public). For public exposures the scope of these limits are restricted
to doses incurred as the result of practices. Doses incurred in situations where the only protective action
takes the form of an intervention are not included in system of dose limits (see action levels). Radon,
thoron and their progeny in open air or in houses, natural radionuclides already in the environment and
cosmic radiation on ground, are examples of those situations.
While these dose limits are called primary limits, for practical reasons further limits (secondary or
derived limits) are specified which are given in terms of operational or other quantities and derived from
the primary limits. They are applied, for example, to define control or prohibited areas or annual limits of
intake (ALI) of radionuclides. The ALI values which are specific for each radionuclide considered are
also based on the committed dose for the same periods as mentioned above.
The primary dose limits internationally recommended by ICRP [91I1] and the IAEA [96IA] are given
below. Many, but not all countries have transferred these values into their national legislation and
regulations. Some countries have either less or more restrictive regulations. As a consequence, the legal
dose limits may, therefore, be different in different countries.
For occupational exposure the effective dose is limited to 20 mSv per year averaged over 5 years (100
mSv in 5 years) with the further provision that the effective dose should not exceed 50 mSv within any
single year. This limit avoids any deterministic effects of exposures and limits the stochastic effects to a
risk level of about 103. For internal exposure the committed dose limit is restricted to 20 mSv in each
year and the annual limit of intake (ALI) is related to this value.
For women, when pregnancy has been declared, the embryo and foetus should be protected by
applying for external exposure an additional equivalent dose limit to the surface of its abdomen of 2 mSv
for the remaining period of the pregnancy and limiting the intake of radionuclides to 1/20 of the annual
limit of intake.
The detriment due to external weakly penetrating radiation mainly concerns the skin or the eye lens.
In order to avoid deterministic effects the skin dose is, therefore, additionally limited to 500 mSv per year
(averaged over any 1 cm2, regardless of the area exposed) and the dose to the eye lens to 150 mSv per
year. For the same reason, the annual equivalent dose to the extremities (hands, feet) is also limited to 500
mSv.
The approach for choosing dose limits to the public may be either based on the same ideas as for
occupational exposure considering, however, the fact of the large number of persons involved or on the
judgement on the existing dose level from natural radiation sources and its variation in different places
where no influence on the health detriment of the population has been observed.
For public exposure from sources given in practices, the ICRP has recommended a limitation of the
effective dose to 1 mSv per year. In special circumstances, however, a higher value may be allowed in a
single year if the average over 5 years does not exceed 1 mSv per year. The ICRP has also defined
additional annual limits for the skin and the eye lenses which are 1/10 of the value recommended for
workers (50 mSv averaged over any 1 cm2 of the skin and 15 mSv for the lens of the eye).

Dose constraints
The control of public exposure in normal situations is usually performed by the application of controls at
the different sources applying procedures of constrained optimisation and the use of prescriptive limits. A
dose constraint, which is a value of individual dose from a defined source, should be used in the
optimisation of protection to exclude protection options that would result in individual doses exceeding
the constraint. Dose constraints are an integral part of the optimisation of protection and are thus
prospectively. They are not, however, limits to be applied retrospectively.

Landolt-Brnstein
New Series VIII/4

Ref. p. 4-27]

4 Radiological quantities and units

4-25

The above mentioned annual limit for occupational exposure means implicitly that the dose constraint
for optimisation should not exceed 20 mSv per year. It is often convenient to define a homogeneous
group of persons a critical group which are assumed to be most highly exposed by the single source
considered and to apply the dose constraint to the mean dose in that critical group.
For medical exposures no dose limits have been recommended because a radiotherapy or diagnostic
treatment should always provide a direct benefit to the patient. The choice of the practice and its
performance should be optimized with respect to applied doses and the medical benefit.
In order to characterize good medical practice and to enable quality assurance programs for use in
these cases, it is helpful to define constraints or reference values based on the actual state of the art of the
various investigations and procedures. Such values are especially given in the various practices of
diagnostics with X-rays and radionuclides.

Action levels
While for the situation of occupational exposures generally dose limits are defined, there are other
situations where the only protective action takes the form of an intervention, e.g. in cases of public
exposure in areas of high level of natural radiation or in areas contaminated because of former human
activities or accidents like e.g. nuclear bomb testing or the Chernobyl accident.
For intervention situations action levels may be defined which specify dose levels or an activity
concentration in air or the specific activity in materials of the environment which are of concern with
respect to public exposure. If such a level is exceeded, this should initiate measures for a reduction of the
exposure and different action levels may define different measures characterising the strength of the
necessary intervention.
While for public exposure from sources given in practices, a dose limit of 1 mSv per year is
recommended which is in the order of the natural background exposure excluding radon, action levels for
initiating protection measures to the public are mostly higher, depending on the strength of the
recommended measures.
A typical case for the definition of action levels is the exposure by radon and its progenies. Radon is
always present in the environment and may appear in higher concentrations at specific work places or in
homes. For radon, however, the specification of a dose level for actions is relatively complex because the
dose coefficient relating a mean radon concentration at a place to an effective dose value depends on the
mean equilibrium factor F (see 4.6.2) and the mean annual time people stay at this place. Furthermore, the
coefficient is mainly based on modelling and includes a large uncertainty (see Chapt. 7). Mostly the radon
action levels are given in terms of Bq m3 or Bq m3 h.
For occupational exposure with the assumption of 2000 h at work and a mean equilibrium factor of
0.4 a conversion factor of 156 Bq m3 mSv1 or 62 Bq m3(EEC)mSv1, respectively, is given [94I1].
Actions for reducing the radon concentration at a work place may be performed if the mean annual radon
concentration is in the range from 500 Bq m3 to 1500 Bq m3 (1000 Bq m3 corresponds to about 6 mSv
per year for 2000 h at work and F = 0.4). A mean annual concentration of 3000 Bq m3 corresponds to
about 20 mSv per year which is equal to the dose limit for occupational exposure.
For public exposure in dwellings a conversion factor of 58 Bq m3 mSv1 or 23 Bq m3(EEC)mSv1,
respectively, is given [94I1] under the assumption that a person stays 7000 h per year in the house (with F
= 0.4 in the house) and the other time in free air (with a low radon concentration). Action levels are
recommended by the ICRP also for this case. They are based on the following ideas. There exists a range
of normal mean radon concentrations in dwellings where no actions are necessary or useful. For existing
houses with higher mean concentrations actions for reducing such values should be considered. Future
houses should be designed to stay within the normal range. Because of the very different situation in the
various regions regarding the natural radon concentration in the ground and hence in houses, local
recommendations or regulations may differ strongly in the different countries. The ICRP has specified an
upper boundary of the normal mean radon concentration in dwellings with 200-400 Bq m3 depending on
the regional situation.
Landolt-Brnstein
New Series VIII/4

4-26

4 Radiological quantities and units

[Ref. p. 4-27

Another situations which may occur are emergencies where people or the government needs advice
what type of actions are necessary under given or expected exposures, e.g. staying at home, avoiding to
eat fresh vegetables or drink fresh milk or leaving a defined area for some time. Such a situation is also a
type of intervention where action levels may be defined in national regulations like those mentioned
above. Principles and more detailed information are given in ICRP Publication 63 [93I4]. For immediate
emergency situations there may also dose values be given for fireman and other rescue personnel in order
to restrict their risks due to an exposure. Dose levels may be defined which should not be exceeded in one
case or annually or in life time.

Landolt-Brnstein
New Series VIII/4

4 Radiological quantities and units

4-27

4.9 References
73Jac
77I1
80I1
85I1
86I1
88I1
91BI
91I1
93I1
93I2
93I3
93I4
93Se
94I1
94I2
96IA
96I1
97I1
98I1
98I2
98NN
99Ch

00I1
01I1

Jacobi, W.: The concept of effective dose - a proposal for the combination of organ doses.
Radiat. Environ. Biophys. 12 (1975) 101.
ICRP: Recommendations of the international commission on radiological protection. ICRP
Publication 26, Ann. ICRP 1 (3) (1977).
ICRU: Radiation quantities and units. ICRU Report 33, Washington, 1980.
ICRU: Determination of dose equivalents resulting from external radiation sources. ICRU
Report 39. Bethesda, MD: ICRU Publications, 1985.
ICRU: The quality factor in radiation protection. ICRU Report 40. Bethesda, MD: ICRU
Publications, 1986.
ICRU: Measurement of dose equivalents from external radiation sources, Part 2. ICRU Report
43. Bethesda, MD: ICRU Publications, 1988.
Bureau International des Poids et Mesures: Le Systme International dUnits (SI). 6th edition.
Pavillon de Breteuil, Sevres, 1991.
ICRP: Recommendations of the international commission on radiological protection. ICRP
Publication 60, Ann. ICRP 21 (1-3) (1991).
ICRU: Quantities and units in radiation protection dosimetry. ICRU Report 51. Bethesda, MD:
ICRU Publications, 1993.
International Organisation for Standardisation: ISO Standards Handbook, Quantities and Units,
3rd edition, Geneva: International Organisation for Standardisation, 1993.
ICRP: Age-dependent doses to members of the public from intake of radionuclides: Part 2
Ingestion Dose Coefficients. ICRP Publication 67, Ann. ICRP 23 (4) (1993).
ICRP: Principles for intervention for protection of the public in a radiological emergency.
ICRP Publication 63, Edited by ICRP, 1993.
Seltzer, S.M.: Calculation of photon mass energy-transfer and mass energy-absorption
coefficients. Radiat. Res. 136 (1993) 147.
ICRP: Protection against Radon-222 at home and at work. ICRP Publication 65, Ann. ICRP 23
(2) (1994).
ICRP: Dose coefficients for intakes of radionuclides by workers. ICRP Publication 68, Ann.
ICRP 24 (4) (1994).
IAEA: International basic safety standards for protection against ionizing radiation an for the
safety of radiation sources. Safety Series No. 115, International Atomic Agency, Vienna, 1996.
ICRP: Conversion coefficients for use in radiological protection against external radiation.
ICRP Publication 74, Ann. ICRP 26 (3-4) (1996).
ICRP: General principles for the radiation protection of workers. ICRP Publication 75, Ann.
ICRP 27 (1) (1997).
ICRU: Fundamental quantities and units. ICRU Report 60. Bethesda, MD: ICRU Publications,
1998.
ICRU: Conversion coefficients for use in radiological protection against external radiation.
ICRU Report 57. Bethesda, MD: ICRU Publications, 1998.
NNDC: Nuclear Data, Decay Radiations. National Nuclear Data Center, Brookhaven National
Laboratory, Upton, NY, 1998.
Chadwick, M.B., Barshall, H.H., Caswell, R.S., DeLuca, P.M., Hale, G.M., Jones, D.T.L.,
MacFarlane, R.E., Meulders, J.P., Schuhmacher, H., Schrewe, U.J., Wambersie, A., Young,
P.G. A.: Consistent set of neutron kerma coefficients from thermal to 150 MeV for biologically
important materials. Med. Phys. 26 (6) (1999) 974.
ICRU: Nuclear data for neutron and proton radiotherapy and for radiation protection. ICRU
Report 63. Bethesda, MD: ICRU Publications, 2000.
ICRU: Determination of operational dose equivalent quantities for neutrons. ICRU Report 66,
J. ICRU 1 (3) (2001).

Landolt-Brnstein
New Series VIII/4

Ref. p. 5-32]

5 Shielding against ionizing radiation

5-1

5 Shielding against ionizing radiation

In the shielding calculation for the radiation facility, simple dose estimation methods by using the
shielding calculation constants are effective and widely used. These shielding calculation constants
depend on the dose quantity to be estimated. Chapter 5 presents simple calculation methods and related
constants for charged particles, photons and neutrons.

5.1 Introduction
Shielding is an essential element of practical radiation protection. The necessary precautions depend
especially on the kind of radiation. Charged particles as electrons and alpha particles can be absorbed in
matter totally, as they have a maximum penetration depth, depending on their charge, mass, energy and
on the properties of the absorbing media. A shielding slab will attenuate photons and neutrons just to a
fraction of their primary intensity on the other hand, without enabling total absorption.
Thus shielding calculations for charged particles mainly concern the evaluation of maximum
penetration depths. As to photons an analytical calculation method has proved successful for simple
geometries. It is based on the exponential law of attenuation of the unscattered photon component and a
build-up portion for the scattered radiation component. The treatment of neutrons is more complicated
than photon calculations. Simple methods to estimate dose rates can only be described for typical neutron
sources and shielding materials, using results of more powerful shielding codes.

5.2 Stopping power and range


To calculate the penetration of charged particles in matter, it is necessary to have information on the basic
interactions that govern the passage through the shield. The predominant effects of protons and alpha
particles are the elastic and inelastic collisions with electrons. While elastic collisions are resulting in a
change of direction for the incident particle, inelastic collisions lead to energy loss and production of
secondary radiation. Electrons traversing some distance in matter lose energy in numerous inelastic
collisions with bound atomic electrons along their track. Furthermore Bremsstrahlung production
becomes important in electron transport especially for high-Z media and high energies.
The basic quantity for shielding purposes of charged particles is the stopping power, which is defined
as the average energy loss per unit path length. It can be separated into the components collision stopping
power Scol due to Coulomb collisions and radiative stopping power Srad due to Bremsstrahlung production.
The range of charged particles is usually estimated on the basis of the continuousslowing
downapproximation (csda). In this approximation particles are assumed to loose their energy
continuously in the course of slowing down, with a fixed energy loss per path length given by the
stopping power. That means energy-loss fluctuations are neglected. Integrating the reciprocal of the total

Landolt-Brnstein
New Series VIII/4

5-2

5 Shielding against ionizing radiation

[Ref. p. 5-32

stopping power with respect to kinetic energy E gives the csda-range r0, which is a rather good
approximation of the mean path length by a particle on its course to rest. Generally the csda-range of a
particle of initial energy E0 slowing down in matter to the rest energy Er is evaluated from the expression
E0

r0 =

[S (E )

col

+ S ( E ) rad ]1 dE

(5.2.1)

Er

Because of the numerous scattering processes with angular deflections the csda-range is in most cases
much larger than its projection on the initial direction of the particle track or the penetration depth. Tables
of stopping powers and csda-ranges for protons and alpha particles are given in ICRU Report 49 [93ICR].
Corresponding values for electrons are presented in ICRU Report 35 [84ICR1], ICRU Report 37
[84ICR2], and ICRU Report 56 [97ICR]. An early review is given by Knop and Paul [64Kno].

5.3 Penetration depths of charged particles


5.3.1 Heavy charged particles
Protons and alpha particles keep their initial direction rather far on their way, except near the end of the
track. Because of this deviation from linear tracks, caused by multiple scattering, the average penetration
depth R is a more useful quantity than the csda-range. It gives the expectation value of the distance in the
initial direction of motion to that point, where the particle has slowed down to rest. The deviation from
linearity of the particle track is described by the detour factor. It is practically equivalent to R/r0. The
detour factor increases with increasing energy and decreasing atomic number. For this reason detour
factors become important for low energies. The values are about 0.8 and 0.65 for protons and alpha
particles of 1 MeV in lead, respectively. Fig. 5.3.1 gives average penetration depths R for common used
materials of density , estimated on the basis of the csda-range and detour factor tables of ICRU Report
49 [93ICR]. Table A5.3.1 gives the corresponding numerical values for some further materials as well
(CD-ROM).
As collision stopping powers at a given particle velocity are the same for all particles with the same
charge number, proton ranges can be used as well for the estimation of the ranges of deuterons and
positively charged pions and muons. At a given velocity the kinetic energy E of a particle with mass m is
related to the kinetic energy Ep of a particle with mass mp by
E=

m
Ep
mp

(5.3.1)

The relation between the range R(E) of particles with charge number z and the proton-range Rp(Ep) is
given by
R( E ) = R p (m p / m E )

m 1
Fcorr
mp z2

(5.3.2)

The correction factor Fcorr can be assumed to be unity for deuterons and positively charged pions and
muons. It takes into account above all uncertainties of the particle charge. It is near unity for light ions at
high particle energies. As slow ions can capture and lose electrons the effective charge may become much
smaller than the nominal charge, resulting in a reduced stopping power. For alpha particles Fcorr
approaches unity above 1 MeV and may increase to about 2 at lower energies. Fig. 5.3.2 gives further
average penetration depths for protons and alpha particles in air on the basis of the csda-approximation.
The average penetration depths for deuterons in air are calculated by Eq. (5.3.2). Table A5.3.2 gives the
corresponding numerical values (CD-ROM).
Landolt-Brnstein
New Series VIII/4

Ref. p. 5-32]

5 Shielding against ionizing radiation

5-3

10

Average penetration depth R [g cm2]

10

lead
-1
10
iron
water
-2
10
aluminum
-3
10

-4
10
0.1

Fig. 5.3.1. Average penetration depth R of protons in


water, aluminium, iron and
lead.
1

10

100

Proton energy E [MeV]

Fig. 5.3.2. Average penetration


depth R of protons, deuterons
and alpha particles in air.
( = 1.205103 g/cm3).

Sample problem
Average penetration depth of 3 MeV tritons in air.
From Eq. (5.3.2) and Fig. 5.3.2 results:
R(3 MeV) = Rp(1/3 3 MeV) 3/1 1/12 = 3 Rp(1 MeV) = 3 2.35 cm = 7.05 cm

Landolt-Brnstein
New Series VIII/4

5-4

5 Shielding against ionizing radiation

[Ref. p. 5-32

5.3.2 Electrons and positrons


The range of electrons depends on the total (collision plus the radiative) stopping power. Monoenergetic
electrons and beta rays suffer similar energy and angle straggling as heavy charged particles while
slowing down. The deflections are however comparatively large because of their small mass. Thus the
range resulting from the continuous-slowing-down-approximation (csda) is always larger than the
projection of the particle track on the initial direction. The detour factor is near unit for low-Z media but
reaches values up to 4 for lead.
Different range definitions are used for practical purposes. In shielding calculations the maximum
range Rmax is the most adequate quantity. It is defined as the depth at which the extrapolation of the tail of
the transmission curve (versus electron beam axis depth) meets the Bremsstrahlung background. A
frequently used numerical expression for the maximum range Rmax of beta rays of maximum beta energy
Emax in matter of density is given by
Rmax = 0.11 + 0.0121 + ( Emax / 1.92) 2

(5.3.3)

where Emax is in MeV and Rmax in g cm2 [97ICR]. The relation is useful in the energy range between
0.05 MeV and 5 MeV, for monoenergetic electrons as well. Fig. 5.3.3 gives maximum ranges for six
common used materials, calculated by Eq. (5.3.3). Table A5.3.3 gives the corresponding numerical values
(CD-ROM).
2

10

Maximum range Rmax [cm]

10

air*

10

glass

water

Al
-1

Fe

10

Pb
-2

10

-3

10

0.1

10

Fig. 5.3.3. Electron ranges


Rmax in air (1.205103), water
(1.0), glass (2.23), Al (2.7), Fe
(7.87) and Pb (11.35). Values
in parentheses: densities in
g/cm3. *Maximum range Rmax
in m.

Electron energy E [MeV]

A rather simple empirical expression for the maximum range of electrons of energy E in matter of
density is given by
Rmax = E/2

(5.3.4)

where E is in MeV and Rmax in g cm2. Maximum ranges evaluated from Eq. (5.3.3) and (5.3.4) are
shown in Fig. 5.3.4 together with csda-ranges for water and lead [84ICR2]. Eq. (5.3.4) turns out to be a
cautious overall approximation of the maximum range overestimating for energies below 0.3 MeV and
above 20 MeV. Eq. (5.3.3) provides considerable overestimation only in the energy range above 20 MeV.
Table A5.3.4 gives the numerical values of the csda-ranges of air, water, Be, Al, Fe, Pb (CD-ROM).
Landolt-Brnstein
New Series VIII/4

Ref. p. 5-32]

5 Shielding against ionizing radiation

5-5

10

Electron range R [gcm2]

water(csda)
lead (csda)

Eq. (5.3.4)

0.1

Eq. (5.3.3)
0.01
0.1

10

100

Fig. 5.3.4. Electron ranges


R evaluated from Eq.
(5.3.3), Eq. (5.3.4) and by
csda-approximation
for
water and lead.

Electron energy E [MeV]

Positrons undergo the same interactions in matter as electrons. Because of differences in collision and
radiative stopping powers the positron csda-range varies between shortening at low energies and
prolongation at high energies. The shortening of the positron csda-range (with respect to the
corresponding electron csda-range) will amount up to 7 % at 0.1 MeV. At energies near 100 MeV the
prolongation will be up to 11 % in lead and up to 2 % in water. On the assumption of positrons being
annihilated before being slowed down to rest a further shortening of the positron range of up to 4 % in
lead has to be considered [84ICR2].
In shielding of beta sources the range curves will yield a sufficient estimate of the necessary slab
thickness. With high source activities Bremsstrahlung resulting from the deceleration of the beta particles
in the material may need to be shielded as well.

5.4 Photons
5.4.1 Basic shielding concept
For most gamma shielding studies photon energies of 10 keV to 10 MeV are important. In this energy
range, the photoelectric effect, pair production and Compton scattering mechanisms of interaction
predominate over all others. Of these three interactions, the photoelectric effect predominates at the lower
photon energies; pair production is important only for higher-energy photons, while Compton scattering
predominates at intermediate energies. In a few cases the shielding analyst may need to account also for
coherent (Rayleigh) scattering, annihilation and fluorescence radiation. Most shielding analysis involves a
study of the fluence field at pertinent locations with respect to the outside or inside shield. The purpose of
such analysis is to predict the corresponding responses of some type of detector, and therefore the field
information must be converted into the detector responses. These relate to the fluence by a multiplier
called the detector response function. The fluence of photons is the quotient of N by a, where N is
the number of photons, which enter a sphere of cross-sectional area a. Detector responses for photons of
interest are exposure, air kerma, absorbed dose and ambient dose equivalent.

Landolt-Brnstein
New Series VIII/4

5-6

5 Shielding against ionizing radiation

[Ref. p. 5-32

In this chapter the ambient dose equivalent is used in accordance with ICRP Report 74 [96ICR]. The
ambient dose equivalent rates are obtained by a shielding code that satisfies the Boltzmann transport
equation. Using these calculated results, a simple method to estimate the ambient dose equivalent rate for
typical radioactive sources and typical shielding media is introduced.

5.4.2 Attenuation data of radioactive sources in shielding materials


The radionuclides dealt with in this chapter are shown in Table 5.4.1. The value of the ambient dose
equivalent rate depends on the photon energy, the geometric configuration of the source, the nature and
the thickness of the shielding material. The photon fluences emitted from a radioactive source with
ordinary concrete ( = 2.10 g/cm3), iron, lead and water as shielding materials are calculated using the
BERMUDA code [92Suz, 93Suz]. Densities and elemental weight fractions of ordinary concrete
( = 2.10 g/cm3 and 2.35 g/cm3) are shown in Table A5.4.1 (CD-ROM). The mass attenuation
coefficients used were taken from the photon library PHOTX [88DLC], which are shown in Table A5.4.2
(CD-ROM).
*
(t ) , where t is the thickness
In this text the ambient dose equivalent rate H& * (10) is represented as H& 10
of the shield layer. The purpose of shielding analysis is to predict the corresponding responses of certain
types of detectors as defined by the symbol R, and to relate R to the fluence (E, t) or to the flux density
(E, t ) of photons of energy E by a multiplier to be called the conversion coefficient; where the flux
density (E, t ) is defined as quotient of the incremental fluence that occurs at a specified position and
the time interval t. The conversion coefficients of the exposure dose rate ( X& / ) E , of the air kerma
rate ( K& / ) and of the ambient dose equivalent rate ( H& * / ) for the flux density [cm2 s1] are given
a

10

in Table A5.4.3 (CD-ROM). While the conversion coefficients of exposure rate ( X& / ) E [R h1 cm2 s]
are taken from the third column of Table A.1 in ICRU Report 47 [92ICR], the ones of the air kerma rate
*
( K& a / ) E [nGy h1 cm2 s] and the ambient dose equivalent rate ( H& 10
/ ) E [nSv h1 cm2 s] are taken from
the fourth and fifth column of Table A.21 in ICRP Report 74 [96ICR]. Here, the three conversion
coefficients are given in special units for convenience sake of calculation.
Consider a point source of activity A [Bq] and a point detector P located at a distance r + t [m] from
the source, as illustrated in Fig. 5.4.1, where a shield layer of thickness t [m] is placed between the source
and the detector. The energy spectrum of the photon flux density at point P is represented by (E, t ). Then
the air kerma rate and ambient dose equivalent rate at point P are represented by the following formulas.

(K& ) (E ,t ) dE
(t ) = (H& ) (E ,t ) dE

K& a (t ) =
*
H& 10

(5.4.1)

*
10

(5.4.2)

By introducing the new constant 10* , the ambient dose equivalent transmission factor T(t), and effective
conversion coefficient f * ( E ,t ) , the ambient dose equivalent rate H& * (t ) is simply obtained.
10

10

*
5.4.2.1 Simple method of calculating the ambient dose equivalent rate H& 10
( t ) for radionuclides
listed in Table 5.4.1

The flux density i of photon energy group i emitted from a radioactive point source of activity A [Bq]
is represented by the following formula, where the detector P is located at distance r [m] from the source.

Landolt-Brnstein
New Series VIII/4

Ref. p. 5-32]

i =

A
4 r 2

5 Shielding against ionizing radiation

5-7

(5.4.3)

Ii

Ii represents the transition yield (number of gamma rays per decay). Then, the ambient dose equivalent
rate is obtained by the following summation.
H& 10* =

( H&

/ ) i i

*
10

(5.4.4)

Constant: 10*
With regard to Eq. (5.4.3) and Eq. (5.4.4), the constant 10* [Sv m2 MBq1 h1] is defined as the sum of
components 10* ,i for specified photon energy groups i of a gamma-emitting nuclide, as given in Eq.
(5.4.5).

10* =

*
10,i

4 I ( H&
i

*
10

/ ) i 0.1

(5.4.5)

That means, the value of 10* represents the ambient dose equivalent rate H& 10* [Sv h1] for A = 1 MBq and
r = 1 m of a gamma-emitting nuclide. The values of 10* for 33 radionuclides are given in Table 5.4.1
[01JRIA]. With the constant 10* the ambient dose equivalent rate of a point source of a gamma-emitting
radionuclide of activity A at distance r becomes
A
H& 10* = 2 10*
r

(5.4.6)

Ambient dose equivalent transmission factor: T(t)


The ambient dose equivalent transmission factor T(t) is the quotient of the ambient dose equivalent rate
*
H& 10
(t ) by the ambient dose equivalent rate in the absence of shielding material H& 10* (0) .
T (t ) = H& 10* (t ) / H& 10* (0)

(5.4.7)

The values of the ambient dose equivalent transmission factor T(t) for 33 radionuclides are given in
Table A5.4.4 through A5.4.36 (CD-ROM) for four shielding materials, namely iron, lead, concrete
( = 2.10 g/cm3) and water [01Sak]. Furthermore, the transmission factors T(t) are presented in Fig. 5.4.2
through 5.38, as a function of the thickness t of shield layers [01Sak].
Using the constant 10* and the ambient dose equivalent transmission factor T(t), the ambient dose
equivalent rate H& * ( t ) at a point detector P located a distance r + t [m] from the source, as illustrated in
10

Fig.5.5, is obtained by Eq.(5.4.8).


H& 10* (t ) =

A
10* T (t )
( r + t )2

Landolt-Brnstein
New Series VIII/4

(5.4.8)

5-8

5 Shielding against ionizing radiation

[Ref. p. 5-32

*
5.4.2.2 Simple calculation of ambient dose equivalent rate H& 10
( t ) for radionuclides unlisted in
Table 5.4.1

Effective (averaged) conversion coefficient: f10* ( E0 ,t )


When f10* ( E ) defines the quotient of conversion coefficient ( H& 10* / ) E by ( K& a / ) E for a specified energy
E, the ambient dose equivalent rate in Eq. (5.4.2) is given by the following formula:

*
H& 10
(t ) =

*
10

(E ) (K& a

)E (E ,t ) dE

(5.4.9)

Equation (5.4.9) averaged by air absorbed dose rate spectrum at the point P in Fig. 5.4.1 gives Eq.
(5.4.10). This formula was introduced as the effective conversion coefficient f10* = f10* ( E0 , t ) for photons
of primary energy E0 and for shield layer thickness t is represented by Tanaka and Suzuki [91Tan]:

*
10

f
=

*
10

(E ) (K& a

)E (E , t ) dE

(5.4.10)

(K& ) (E ,t ) dE
a

Then, the relationship between the ambient dose equivalent rate and the air kerma rate becomes
*
H& 10
( t ) = f 10* K& a (t )

(5.4.11)

Effective conversion coefficients f10* = f10* ( E0 , t ) for iron, lead, concrete and water are given in
Table A5.4.37 through A5.4.40 for photon energies from 15 keV to 10 MeV and shield layers up to 40
mfp (CD-ROM) [01Sak].

*
5.4.2.3 Calculation method of ambient dose equivalent rate H& 10 ( t ) using exposure dose rate and
effective conversion coefficient

Step 1: The flux density of uncollided photons of energy E0 for source intensity S = A I at distance r + t
behind a shield layer of thickness t is

0 (E0 ) =

S
exp( t )
2
4 (r + t )

(5.4.12)

The mass attenuation coefficients are listed in Table A5.4.2 (CD-ROM).


Step 2: The exposure dose rate for uncollided photons is

X& 0 (E0 ) = 0 (E0 ) X&

E0

(5.4.13)

The conversion coefficients ( X& / )E are listed in Table A5.4.3 (CD-ROM).


Step 3: The total exposure dose rate including collided photons is
X& = B X& 0

(5.4.14)

Landolt-Brnstein
New Series VIII/4

Ref. p. 5-32]

5 Shielding against ionizing radiation

5-9

Exposure buildup factors B = B(E0, t ) for a point isotropic source in iron, lead, concrete ( = 2.35 g/cm3)
and water are given in Table A5.4.41 through A5.4.44 for photon energies from 15 keV to 10 MeV and
shield layers up to 40 mfp (CD-ROM), which are taken from ANSI/ANS6-4-3 [91ANS].
Step 4: The air kerma rate obtained from the total exposure dose rate is
K& a = 8.764 10 3 X&

(5.4.15)

where 8.764103 is the value of a typical conversion coefficient from exposure to air kerma, X& is in
R h1 and in Gy h1.
Step 5: Using the effective conversion coefficient f10* the ambient dose equivalent rate follows from
Eq. (5.4.11).
Values for not tabulated energies are obtained by interpolation.

5.4.3 An example of the calculation of an ambient dose equivalent rate


Sample problem: 60Co source of A = 3.71013 Bq (1,000 Ci) is treated in a room enclosed by concrete
wall of 100 cm thickness. Calculate the ambient dose equivalent rate at a point P on the outside wall.
The calculation model is illustrated in Fig. 5.4.1, where the distance r between the source and the front
of the concrete wall is 500 cm.

5.4.3.1 The method using the constant 10* and ambient dose equivalent transmission factor T(t )
Using the value 10* of 60Co in Table 5.4.1 and the one of T(t) at concrete 100 cm in Table A5.4.11
(CD-ROM), the ambient dose equivalent rate in Eq.(5.4.8) is obtained, as follows.
3.7 10 7 10* T (t )
*
(t ) =
= 1.028 0.354 1.55104 Sv h1 = 56.4 Sv h1
Eq. (5.4.8): H& 10
2
(5 + 1)

Values for not tabulated thicknesses of the shield layer are obtained by linear interpolation of
log T(t ) t [cm].

5.4.3.2 The method using the effective conversion coefficient f10* and the exposure dose rate
conversion coefficient ( X& / )
Simplifying assumption: 60Co emits two gamma rays of E0 = 1.25 MeV per disintegration.
Step 1: The photon flux density for the uncollided photons is:
Eq. (5.4.12):

0 (E0 ) =

3.7 1013 2 exp( 12.02 )


cm2 s1 = 98.56 cm2 s1
4 3.14 6002

The linear attenuation coefficient of concrete ( = 2.10 g/cm3) for 1.25 MeV photons is = 0.1202 cm1,
which is obtained by the linear interpolation of log log E, using the concrete mass attenuation
coefficients of E0 = 1 MeV and 1.5 MeV given in Table A5.4.2 (CD-ROM). With t = 100 cm results
t = 12.02.

Landolt-Brnstein
New Series VIII/4

5-10

5 Shielding against ionizing radiation

[Ref. p. 5-32

Step 2: Calculation of the exposure dose rate of the uncollided photons:

X& 0 (E0 ) = 0 (E0 ) X&

Eq. (5.4.13):

E0

= 98.56 2.182 106 R h1 = 2.151104 R h1

The conversion factor ( X& / ) E for 1.25 MeV is obtained by linear interpolation of log ( X& / ) E log E,
using ( X& / ) for 1 MeV and 1.5 MeV given in Table A5.4.3 (CD-ROM).
E

Step 3: Calculation of total exposure dose rate including collided photons:


X& = B X& 0 = 21.84 2.151 104 R h1 4.698103 R h1

Eq. (5.4.14):

The exposure buildup factor for E0 = 1.25 MeV in infinite concrete material is obtained by the following
procedure. At first, concrete buildup factors B = 27.01 and 18.36 are obtained for E0 = 1 MeV and 1.5
MeV for t = 12.02 mfp. They result from the linear interpolation of log B(E) t [mfp], using buildup
factors for t = 10 and 15 mfp from Table A5.4.43 (CD-ROM). Next, buildup factor B = 21.84 is
obtained for E0 = 1.25 MeV by the linear interpolation of log B(E) log E, using buildup factors of E0 = 1
and 1.5 MeV at t = 12.02.
Step 4: Conversion from exposure dose rate X& to air kerma rate K& a :
K& a = 8.764 10 3 4.698 10 3 Gy h1 = 4.117105 Gy h1

Eq. (5.4.15):

*
Step 5: Conversion from air kerma rate K& a to ambient dose equivalent H& 10
(t ):

The effective conversion coefficient f 10* in Eq. (5.4.11) is obtained by the following procedure. At first,
the effective conversion coefficients of concrete f 10* = 1.308 and 1.256 are obtained for E0 = 1 MeV and
1.5 MeV and for t = 12.02. They result from the linear interpolation of log f 10* t [mfp], using f 10* for

t = 10 and 15 mfp from Table A5.4.39 (CD-ROM). Next, f 10* = 1.279 is obtained for E0 = 1.25 MeV by
the linear interpolation of log f 10* log E, using effective conversion coefficients of E0 = 1 and 1.5 MeV at
t = 12.02.
H& 10* (t ) = f 10* K& a = 1.279 4.117 105 Sv h1 = 52.7 Sv h1

Eq. (5.4.11):

*
The values of H& 10
(t ) obtained by the methods in 5.4.3.1 and 5.4.3.2 agree within 7 %.

Table 5.4.1 Constant 10* [Sv m2 MBq1 h1]


Radionuclide
18

F
Na
51
Cr
54
Mn
59
Fe
56
Co
57
Co
60
Co
64
Cu
24

10*

Radionuclide

0.166
0.492
0.00547
0.130
0.171
0.492
0.0206
0.354
0.0307

65

Zn
Ga
68
Ge*
75
Se
81
Rb*
85
Kr
85
Sr
99
Mo*
99m
Tc
67

10*

Radionuclide

0.0847
0.0268
0.158
0.0660
0.104
0.00037
0.0826
0.0444
0.0214

103

Pd*
Ag
111
In
124
Sb
123
I
125
I
131
I
133
Xe
137
Cs*
110m

10*

Radionuclide

10*

0.0254
0.416
0.0663
0.266
0.0288
0.0357
0.0650
0.0170
0.0927

192

0.139
0.0685
0.0193
0.0175
0.251
0.00529

Ir
Au
197
Hg
201
Tl
226
Ra*
241
Am
198

1) Radionuclides attached the mark * includes the effects of daughters radiation equilibrium. As the daughter of
only considered.
2)

10*

does not include effects by photon energy of below 30 keV and emission ratio within 0.1 %. However

226

Ra,

125

222

I and

Rn is
103

Pd

include the effect by photon energy of above 10 keV.

Landolt-Brnstein
New Series VIII/4

Ref. p. 5-32]





5 Shielding against ionizing radiation

5-11

,Q)LJVWRIROORZLQJV\PEROVDUHXVHG
77UDQVPLVVLRQIDFWRU
WOD\HUWKLFNQHVVRIZDWHUDQGFRQFUHWH
WOD\HUWKLFNQHVVRILURQDQGOHDG


t1
2 0
0
10

10

80

120

160

18

-1

10

-2

10

-3

200

10

3

-4

Concrete

10

-5

10

-6

10

Iron

-7

W

20

40

60

80

100

t2

t1
160

240

320

t1

cm
400

0
2
0
10

2
0
10

10
10

24

-1

Na

-2

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

30

60

90

-4

10

-5

150

cm
180

Cr

Water

10

-3

120

51

Water

10

120
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU)VRXUFH

)LJ $SRLQWLVRWURSLFVRXUFH
HQFORVHGE\FRQFUHWHZDOO

80

Water

Lead

cm
240

U
6

40

Concrete

Concrete

Lead

Lead

10
10

-6

Iron

-7

30

60

90

t2

120

150
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU1DVRXUFH

Landolt-Brnstein
New Series VIII/4

Iron

10

20

30

t2

40

50

60
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU&UVRXUFH

5-12

5 Shielding against ionizing radiation

t1
0

120

180

240

t1

cm
300

-1

10

-2

10

-3

10

-4

10

-5

50

100

150

200

cm
250

10
0

10

54

Mn

Water


T

10

60

[Ref. p. 5-32

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

59

Fe

Water

Concrete

Concrete
Lead

Iron

Iron

10

-6

10

-7

Lead

20

40

60

80

t2

100
cm

10

10

180

240

300

10
10
10

100
cm

30

60

90

120

150

cm
180

10
0

56

-1

10

Co

-2

10

Water
-3

10

-4

10

Concrete
-5

Lead

10
Iron

-6

10

-7

20

40

60

80

100

t2

10

120
cm

t1
0

60

57

-1

120

180

240

-2

-3

Water

60

-4

Concrete
-5

-6

Lead
Iron

-7

10

20

30

40

50

60
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU&RVRXUFH
t1

cm
300

2 0
0
10

2
0
10
-1

Co

t2

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU&RVRXUFH

10

80

t1

cm
360

10

60

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU)HVRXUFH

T

10

t1

120

40

10

60

20

t2

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU0QVRXUFH

10

Co

25

50

75

cm
100 125 150

64

-1

Cu

10

-2

10

-3

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

Water

Water

10

-4

10

-5

Concrete

Concret e

Lead
Iron

Iron

10

-6

10

-7

Le ad

30

60

90

t2

120

150
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU&RVRXUFH

10

20

30

t2

40

50

60
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU&XVRXUFH

|UQVWHLQ

Landolt-Brnstein
New Series VIII/4

Ref. p. 5-32]

5 Shielding against ionizing radiation


t1

40

80

120

160

200

t1

cm
240

2
0
10

2
0
10

10

-1

10

-2

65

5-13

Zn

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

25

50

75

100

125

67

cm
150

Ga

Water

Water

-3

T

10

10

-4

10

-5

Concrete

Concrete
Iron

10

-6

10

-7

Lead

20

40

60

80

100

t2

120
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU=QVRXUFH
t1
0

50

100

150

200

68

-1

10

-3

68

10

10

-6

10

-7

Water

60
cm

30

60

90

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

120

cm
150

Se

Water

Concrete

Iron

20

40

60

80

100
cm

t2

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU*H*DVRXUFH
t1
30

60

90

Rb-

81m

Kr

10

20

30

40

50
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU6HVRXUFH
t1

cm
120 150 180

81

-1

t2

2
0
10

2
0
10
10

50

Lead

Iron

40

75

Lead

30

-1

Concrete
-5

20

t1
2 0
0
10

Ge- Ga

-4

10

10

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU*DVRXUFH

cm
250

10

-2

T

10

Iron

t2

2
0
10
10

Lead

10

30

60

90

120

85

-1

cm
150

Kr

Water

10

Water
-3

10

10

-2

Concre te

-3

10

-2

10

-4

10

Concr ete

-4

Le ad

Lead

10

-5

10
Iron

10
10

-6

10

-7

10

20

30

t2

40

50

60
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU5EP.UVRXUFH

Landolt-Brnstein
New Series VIII/4

10

-5

Iron
-6

-7

10

20

30

t2

40

50
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU.UVRXUFH

5-14

5 Shielding against ionizing radiation

t1
0

30

60

90

120

150

t1

cm
180

2
0
10

-1

10

-2

Sr

10

-1

10

-2

10

-3

30

60

90

120

99

150

Mo-

99m

cm
180

Tc

Water

Water
-3

10

-4

10

-5

10

-6

10

85

2
0
10

10

[Ref. p. 5-32

Concrete

Concrete

10

-4

Lead

10

Lead

-5

Iron

Iron

10

-7

10

20

30

40

50

t2

60
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU6UVRXUFH
t1
0

10

20

30

2
0
10
10

40

50

10

-6

10

-7

10

20

30

40

50

t2

60
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU0RP7FVRXUFH
t1

cm
60

2 0
0
10

Water

-1

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

20

40

60

80

103

100

Pd-

103m

cm
120

Rh

Concrete
-2

10

-3

10

-4

10

-5

99m

Tc


T

10

Water

Iron

Iron

Concrete

Lead

10
10

-6

-7

10

t2

12
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRUP7FVRXUFH
t1
0

50

100

150

200

10

110m

-1

10

20

30

40

50

t2

60
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU3G5KVRXUFH
t1

cm
250

2
0
10

2
0
10

10

Lead

Ag

-2

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

20

40

60

cm
100 120

80

111

Water

In

-3

10

Water

10

-4

10

-5

10

-6

10

-7

Concrete

Concret e
Lead
Iro n

20

40

60

t2

80

100
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRUP$JVRXUFH

10

Iron
Lead

-7

10

15

t2

20

25

30
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU,QVRXUFH

Landolt-Brnstein
New Series VIII/4

Ref. p. 5-32]

5 Shielding against ionizing radiation


t1

60

120

180

240

t1

cm
300

2 0
0
10

2
0
10

10

124

-1

5-15

Sb

10

30

60

90

120

123

-1

cm
150

Water

10

-2

10

-3

10

-2

10

-3

10

-4

10

-5

10

-6

Wate r

10

-4

10

-5

Concrete

Lead

Concrete
Iron

Lead

10

-6

10

-7

Iro n

20

40

60

80

t2

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU6EVRXUFH
t1
0

10

100
cm

-7

10

20

30

t1

cm
10

2
0
10

20

40

60

Water
-1

10

-2

10

-3

10

Water

10
125

10

Concrete

-4

50
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU,VRXUFH

2
0
10
10

40

t2

10
10

-1

80

131

cm
100

-2

Concrete

-3

-4

Lead
Iron

10

-5

10

Lead

-5

Iron

10

-6

10

-7

10 0.0

0.1

0.2

0.3

0.4

t2

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU,VRXUFH
t1
0

20

40

60

80

10

0.5
cm

133

-1

2
0
10
10

Xe

10

Concrete
-4

10
Lead

-5

10

-6

10

-7

24

32

40
cm

30

60

90

cm
120 150 180

137

-1

Cs

-2

-3

Concrete

10

-3

10

16

W ater

10

t1

cm
100

-2

10

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU,VRXUFH

Water

10

-7

t2

2
0
10
10

-6

-4

Iron

Iron

10

-5

Lead

12

18

t2

24

30
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU;HVRXUFH

Landolt-Brnstein
New Series VIII/4

10

-6

10

-7

10

20

30

t2

40

50

60
cm

)LJ$PELHQWGRVHHTXLYDOHQWWUDQVPLVVLRQ
IDFWRURIVKLHOGVIRU&VVRXUFH

5-16

5 Shielding against ionizing radiation

[Ref. p. 5-32

5.5 Neutrons
5.5.1 Basic shielding concepts
In passing through shielding material, neutrons attenuate by elastic scattering, inelastic scattering and
absorption. For high-energy neutrons over 10 MeV, inelastic scattering reaction is effective to decrease
energy. Therefore iron is a suitable material because of its relatively big inelastic cross section. For
neutrons with energy lower than 10 MeV, materials that contain hydrogen are used to utilize its elastic
scattering reaction and absorption process.
Dose calculations of neutrons are more complicated than photons because secondary gamma-ray dose
by neutron capture and inelastic scattering should be taken into account. Moreover, reaction type and
cross section depends strongly on the neutron energy and the shielding material.
Therefore, dose evaluation is generally done by using a shielding code that solves the Boltzmann
transport equation. In the present chapter a simple method to estimate dose rates from typical neutron
sources and shielding materials is described using results of the shielding code ANISN [73Eng].

5.5.2 Attenuation data of various neutron sources in shield material


Fig. 5.5.1 shows the geometry of the transmission calculations by ANISN. A neutron point isotropic
source with source intensity S = 1 s1 is surrounded by a thick spherical shield.
The JENDL3.2 [90Shi] cross-section library is used in this calculation. The elemental composition
and the density of each shield material are shown in Table 5.5.1. Dose rates were estimated using the
calculated neutron and secondary gamma-ray flux and the flux-to-dose conversion factor of ambient dose
equivalent H (10) in ICRP Report 74 [96ICR].
Table 5.5.3 - 5.5.6 give the calculated neutron and secondary gamma-ray ambient dose rates for 252Cf,
Am-Be, D-D and D-T sources [01Sak]. The first column shows the distance r [cm] from the centre, the
second column shows the equivalent dose rate at distance r when no shield material is present: H& 0 [Sv
h1], the third to fifth columns show the neutron, secondary gamma-ray and total equivalent dose rates in
water at distance r: H& n [Sv h1], H& g [Sv h1] and H& t [Sv h1], and the sixth to eighth columns show
the neutron, secondary gamma-ray, and total transmission factors: Fn (= H& n / H& 0 ), Fg (= H& g / H& 0 ) and Ft
(= H& t / H& 0 ) for water. Fn, Fg, Ft values for polyethylene, ordinary concrete and heavy concrete are also
described. As for the simple shielding estimation, the constant for various sources is shown in Table
5.5.2, representing the bare ambient dose equivalent rate at 1 m for unit source intensity.
Similar tables for the effective dose rates for AP (anterior - posterior) exposure geometry are shown in
Table A5.5.1 - A5.5.4 [01Sak] of the accompanying CD-ROM.
Neutron and secondary gamma-ray ambient dose equivalent rates and effective dose rates are
calculated by the following equation.

Landolt-Brnstein
New Series VIII/4

Ref. p. 5-32]

5 Shielding against ionizing radiation

5-17

S
H& = F (t ) 2
d

(5.5.1)

constant [Sv h1 m2 s] (Table 5.5.2 and A5.5.5 (CD-ROM))


(bare ambient dose equivalent rate or effective dose rate at 1m for unit source intensity)
F(t ) transmission factor (Fn, Fg, Ft ) for layer thickness t (Table 5.5.3 - 5.5.6 and A5.5.1 - A5.5.4,
Fig. 5.5.2 - 5.5.17 and 5.5.20 - 5.5.35 (appendix))
S
neutron source intensity [s1]
d
distance from source to detector [m]
H& Here H& means ambient dose equivalent rate for table 5.5.3 - 5.5.6, and effective dose rate for table
A5.5.1-A5.5.4

Fig. 5.5.2 - 5.5.5 show the neutron and secondary gamma-ray dose transmission curves for a 252Cf
source in water, polyethylene, ordinary concrete, and heavy concrete. As shown in these figures,
secondary gamma-rays play dominant role from about 50 cm in water and polyethylene, and about 150
cm in ordinary concrete. The total transmission factor for heavy concrete is lower than that for the other
materials, because it suppresses secondary gamma-rays efficiently.
Fig. 5.5.6 - 5.5.9 show the transmission curves for Am-Be source in each shield material. The neutron
source spectrum from (, n)-sources depends on the grain radius and mixing ratio. This transmission
factor was calculated using the source spectrum measured by Greiss [68Gre]. Transmission factors for
Am-Be are similar to these for 252Cf source.
Fig. 5.5.10 - 5.5.13 show the transmission curves for D-D source in each shield material. The
transmission factor is low because of the low source neutron energy (2.45 MeV). D(d, p)T and D(d, n)3He
reactions occur with almost the same probabilities. Therefore the ordinary D-D source produces a small
amount of D-T neutrons by accumulated tritium. The D-T neutrons must be considered simultaneously.
Fig. 5.5.14 - 5.5.17 show the transmission curves for D-T source in each shield material. The
transmission factor is very high for D-T neutrons because of the neutron energy being high (14.1MeV).
Similar figures for the effective dose rate for AP (anterior - posterior) exposure geometry are shown in
Fig. 5.5.20 - 5.5.35 of the accompanying CD-ROM.
These attenuation calculations were done in sufficiently thick material, called infinite geometry. This
means, calculated dose rates at every point contain the backscattered components. This causes unrealistic
results, so that dose attenuation factor exceeds one. Backscattered dose contributions depend on the shield
material and thickness. Attention has to be paid as well to a more realistic geometry, which usually shows
a certain distance between the source and the shield wall. This effect increases the transmission factor to a
certain extent.
Considering these effects, overestimation results at 0.5 m to about 1.4 for water and to about 1.2 for
ordinary concrete. Although the attenuation factor is conservative, it is sufficient for easy evaluation.
Table 5.5.1 Elemental composition and densities of shield material. *) Type 02-a concrete from ANL5800, p.660 (1963); **) From JAERI-M 6928, p.36 (1977).
Material
3

Density [g cm ]
Element
H
C
O
Mg
Al
Si
Ca
Fe
Landolt-Brnstein
New Series VIII/4

Water
Polyethylene
1.0
0.93
Atomic densities [1024 cm3]
6.6738102
7.9793102
3.9930102
3.3370102

Ordinary concrete*
2.1

Heavy concrete**
3.715

1.2985102
1.0821104
4.3051102
1.1614104
1.6321103
1.5582102
1.4086103
3.2353104

9.9885103
4.3899102
4.3265105
1.6502104
3.7264103
2.2782103
2.3602102

5-18

5 Shielding against ionizing radiation

[Ref. p. 5-32

Table 5.5.2 Constant (ambient dose equivalent) for various neutron sources
Source
252

Cf
Am-Be
D-D
D-T

[Sv h1 m2 s]
1.11105
1.13105
1.19105
1.49105

FP



U


VRXUFH

 VKLHOGPDWHULDO



)LJ*HRPHWU\IRUWUDQVPLVVLRQFDOFXODWLRQ


G FP




 &I DLU W FP


LURQ
 SRO\HWK\OHQH
W FP





)LJ &IVWRUDJHFRQWDLQHU

G FP



&I

FP
DLU

FRQFUHWH
W FP

)LJ&IH[SRVXUHURRP

5.5.3 Sample shield calculation


Sample problem: 252Cf neutron source of 37 MBq (1.88 g: S = 4.32106 s1) is stored in a storage
container that can be modelled as Fig. 5.5.18. The same bare source is used in a room that can be
modelled as Fig. 5.5.19. Calculate ambient dose equivalent rates at the container surface and outside the
room wall.

Landolt-Brnstein
New Series VIII/4

Ref. p. 5-32]

5 Shielding against ionizing radiation

5-19

(1) Dose calculation at the surface of storage container


Neutron dose rate
The shielding effect by the 0.1 cm thick iron layer is small, therefore the iron is neglected, and
polyethylene is the only shielding material to be considered. With from Table 5.5.2 and the neutron
transmission factor Fn(40) for polyethylene from Table 5.5.3 (t r = 40 cm) the neutron ambient dose
equivalent rate is
4.32 10 6
Eq. (5.5.1): H& n = 1.11105 5.74103
Sv h1 = 1.10 Sv h1
0.52
Secondary gamma-ray dose rate
The shielding effect by the 0.1 cm thick iron layer is small, therefore the iron is neglected, and
polyethylene is the only shielding material to be considered. With from Table 5.5.2 and the secondary
gamma-ray transmission factor Fg(40) for polyethylene from Table 5.5.3 (t r = 40 cm) the secondary
gamma ambient dose equivalent rate is
4.32 10 6
Eq. (5.5.1): H& g = 1.11105 1.26102
Sv h1 = 2.42 Sv h1
0.52

Also, the primary gamma-rays of 252Cf source should be considered.


(2) Dose calculation outside the exposure room
Neutron dose rate
With from Table 5.5.2 and the neutron transmission factor Fn(50) for ordinary concrete from Table
5.5.3 (t r = 50 cm) the neutron ambient dose equivalent rate is
4.32 10 6
Sv h1 = 0.299 Sv h1
Eq. (5.5.1): H& n = 1.11105 1.56101
5.0 2

Secondary gamma-ray dose rate


With from Table 5.5.2 and the secondary gamma-ray transmission factor Fg(50) for ordinary concrete
from Table 5.5.3 (t r = 50 cm) the secondary gamma-ray ambient dose equivalent rate is
4.32 10 6
Sv h1 = 0.0313 Sv h1
Eq. (5.5.1): H& g = 1.11105 1.63102
5.0 2

5.5.4 Induced activity


Structure materials, air, coolant waters etc. are activated in neutron fields. The induced activity has to be
considered in radiation protection design of nuclear reactors, fusion experimental reactors and high
energy accelerators. Examples of well-known activation reactions, half-lives, and gamma-ray energies of
produced nuclides typical for nuclear reactors are listed in Table 5.5.7.
These reaction cross sections strongly depend on neutron energy. The induced activity and gamma-ray
dose rate can be estimated by the following three steps:
1) Estimation of the neutron energy spectrum with a computer code such as ANISN [73Eng].
2) Estimation of the induced activity and gamma-ray source strength for a given irradiation and decay
time with a computer code such as ORIGEN [73Bel], using the neutron energy spectrum data
3) Estimation of the gamma-ray dose at a given point with a computer code such as ANISN.

Landolt-Brnstein
New Series VIII/4














































(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

U>FP@

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+Q

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+J

)Q

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

:DWHU

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+W
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

)J
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

)W
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

3RO\HWK\OHQH
)J
)W

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

)Q

Table 5.5.3 Neutron and secondary gamma-ray ambient dose equivalent rates for 252Cf

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

2UGLQDU\&RQFUHWH
)Q
)J
)W
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+HDY\&RQFUHWH
)J
)W

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

)Q

5-20
5 Shielding against ionizing radiation
[Ref. p. 5-32

Landolt-Brnstein
New Series VIII/4














































(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

U>FP@

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+Q

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+J

)Q

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

:DWHU

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+W
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

)J
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

)W
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

3RO\HWK\OHQH
)J
)W

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

)Q

Table 5.5.4 Neutron and secondary gamma-ray ambient dose equivalent rates for Am-Be

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

2UGLQDU\&RQFUHWH
)Q
)J
)W
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+HDY\&RQFUHWH
)J
)W

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

)Q

Ref. p. 5-32]

Landolt-Brnstein
New Series VIII/4

5 Shielding against ionizing radiation


5-21














































(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

U>FP@

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+Q

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+J

)Q

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

:DWHU

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+W
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

)J
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

)W
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

3RO\HWK\OHQH
)J
)W

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

)Q

Table 5.5.5 Neutron and secondary gamma-ray ambient dose equivalent rates for D-D

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

2UGLQDU\&RQFUHWH
)Q
)J
)W
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+HDY\&RQFUHWH
)J
)W

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

)Q

5-22
5 Shielding against ionizing radiation
[Ref. p. 5-32

Landolt-Brnstein
New Series VIII/4














































(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

U>FP@

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+Q

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+J

)Q

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

:DWHU

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+W
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

)J
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

)W
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

3RO\HWK\OHQH
)J
)W

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

)Q

Table 5.5.6 Neutron and secondary gamma-ray ambient dose equivalent rates for D-T

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

2UGLQDU\&RQFUHWH
)Q
)J
)W
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

+HDY\&RQFUHWH
)J
)W

(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(
(

)Q

Ref. p. 5-32]

Landolt-Brnstein
New Series VIII/4

5 Shielding against ionizing radiation


5-23

5-24

5 Shielding against ionizing radiation

[Ref. p. 5-32

Table 5.5.7 Typical activation reactions and cross section data, half-lives, gamma-ray energies of
produced radionuclides [81Mug], [88Mcl], [02Nak].
Material
Stainless
steel
Water

Reaction
Ni(n, p)58Co

Half life
70.8 d

-ray energy [MeV]


0.811, 0.511

54

312 d

0.835

59

Co(n, )60Co
16
O(n, p)16N

5.27 y

1.17, 1.33

8.2102 (Fission spectrum averaged)


3.7101 (at 0.0253 eV)

7.13 s

6.13

2.0105 (Fission spectrum averaged)

40

1.83 h

1.29

6.6101 (at 0.0253 eV)

58

Fe(n, p)54Mn

Ar(n, )41Ar

Air
10

Cross section [barn]


1.1101 (Fission spectrum averaged)

10
252

252

Cf/water

Cf/Polyethylene

10

10

10

Transmission factor

Transmission factor

10

10

10

Fn
Fg
Ft

10

10

Fn
Fg
Ft

10

11

10

10

11

50

100
150
Radius [cm]

200

250

Fig. 5.5.2. Transmission factor of water for 252Cf source.

10

50

100
150
Radius [cm]

200

250

Fig. 5.5.3. Transmission factor of polyethylene for 252Cf


source.

10

10
252

252

Cf/ordinary concrete

Cf/heavy concrete

1
1

10

Transmission factor

Transmission factor

10

10

10

Fn
Fg
Ft

10

10

10

10

Fn
Fg
Ft

10

10

10

10

50

100
150
Radius [cm]

200

250

Fig. 5.5.4. Transmission factor of ordinary concrete for


252
Cf source.

10

50

100
150
Radius [cm]

200

250

Fig. 5.5.5. Transmission factor of heavy concrete for


252
Cf source.

Landolt-Brnstein
New Series VIII/4

Ref. p. 5-32]

5 Shielding against ionizing radiation


10

10

Am-Be/water

Am-Be/Polyethylene
1

10

Transmission factor

Transmission factor

10

10

10

Fn
Fg
Ft

10

10

10

10

Fn
Fg
Ft

10

10

10

5-25

11

50

100
150
Radius [cm]

200

250

Fig. 5.5.6. Transmission factor of water for Am-Be


source.

10

50

100
150
Radius [cm]

200

250

Fig. 5.5.7. Transmission factor of polyethylene for


Am-Be source.

10

10

Am-Be/heavy concrete

Am-Be/ordinary concrete
1

1
1
2

Transmission factor

Transmission factor

10

10

10

10

Fn
Fg
Ft

10

10

10

Fn
Fg
Ft

10

10

10

10

50

100
150
Radius [cm]

200

250

Fig. 5.5.8. Transmission factor of ordinary concrete for


Am-Be source.

Landolt-Brnstein
New Series VIII/4

10

50

100
150
Radius [cm]

200

250

Fig. 5.5.9. Transmission factor of heavy concrete for


Am-Be source.

5-26

5 Shielding against ionizing radiation

10

[Ref. p. 5-32

D-D/water

10

D-D/Polyethylene

10

10

10

10
10

Transmission factor

Transmission factor

5
7

10

10

10

10

12

10

10

14

10

11

10

16

10

Fn
Fg
Ft

13

10

15

10

10

20

10

17

10

Fn
Fg
Ft

18

22

50

100
150
Radius [cm]

200

250

Fig. 5.5.10. Transmission factor of water for D-D


source.

10

50

100
150
Radius [cm]

200

250

Fig. 5.5.11. Transmission factor of polyethylene for


D-D source.

10

10

D-D/heavy concrete

D-D/ordinary concrete

1
1

10

Transmission factor

Transmission factor

10

10

10

Fn
Fg
Ft

10

10

10

10

Fn
Fg
Ft

10

10

10

10

50

100
150
Radius [cm]

200

250

Fig. 5.5.12. Transmission factor of ordinary concrete for


D-D source.

10

50

100
150
Radius [cm]

200

250

Fig. 5.5.13. Transmission factor of heavy concrete for


D-D source.

Landolt-Brnstein
New Series VIII/4

Ref. p. 5-32]

5 Shielding against ionizing radiation

5-27

10

D-T/water

D-T/Polyethylene

10

Transmission factor

Transmission factor

10

10

10

10

Fn
Fg
Ft

10

10

10

10

Fn
Fg
Ft

10

10

50

100
150
Radius [cm]

200

250

Fig. 5.5.14. Transmission factor of water for D-T


source.

50

100
150
Radius [cm]

200

250

Fig. 5.5.15. Transmission factor of polyethylene for D-T


source.

10

D-T/heavy concrete

D-T/ordinary concrete
1

10
1

Transmission factor

Transmission factor

10

10

10

10

10

10

Fn
Fg
Ft

10

10

10

Fn
Fg
Ft

10

50

100
150
Radius [cm]

200

250

Fig. 5.5.16. Transmission factor of ordinary concrete for


D-T source.

Landolt-Brnstein
New Series VIII/4

50

100
150
Radius [cm]

200

250

Fig. 5.5.17. Transmission factor of heavy concrete for


D-T source.

5-28

5 Shielding against ionizing radiation

[Ref. p. 5-32

5.6 Computer codes and online nuclear data services


Table 5.6.1 gives a collection of typical computer codes for transport calculations of gamma-rays,
neutrons and charged particles with regard to shielding problems. The programs are collected and
distributed by different Data Centres, as for example NEA or NNDC. The necessary data in specified
formats for program testing and evaluation are provided by the centres as well. Some Online Nuclear
Data Services for basic nuclear data and evaluated nuclear data are listed in Table 5.6.2.
Table 5.6.1 Computer codes for shielding and source calculations
Computer Codes
ANISN

Engle Jr., W.W.: A Users Manual for ANISN, A One Dimensional Discrete
Ordinates Transport Code with Anisotropic Scattering. K-1693, 1973

BERMUDA

Suzuki, T. et al.: Development of: A Radiation Transport Code System Part I.


Neutron Transport Codes, JAERI 1327, JAERI, 1992
A Radiation Transport Code System Part II. Gamma Rays Transport Codes.
JAERI-M 93-143, JAERI, 1993

DOORS

DOORS includes:
TORT Three-dimensional neutron/photon transport
DORT Two-dimensional neutron/photon transport
ANISN One-dimensional neutron/photon transport
Rhoades, W.A., D.B. Simpson: The TORT Three-Dimensional Discrete
Ordinates Neutron/Photon Transport Code. ORNL/TM-13221, 1997

DOT-4.2

Rhoades, W.A., D.B. Simpson, R.L. Childs, W.W. Engle Jr.: The DOT-4
Two Dimensional, Discrete-Ordinates Transport Code with Space-Dependent
Mesh and Quadrature. ORNL/TM-6529, 1978

DUCT-III

Tayama, R., H. Nakano, H. Handa, K. Hayashi, H. Hirayama, K. Shin, F.


Masukawa, H. Nakashima, N. Sasamoto: DUCT-III, A Simple Design Code
for Duct-Streaming Radiations. KEK Internal 2001-8, 2001

EGS4

Nelson, W.R., H. Hirayama, W.O. Rogers: The EGS4 Code System. SLAC265, 1985
(Electron Photon Shower Simulation by Monte-Carlo)

ETRAN

Berger, M.J., S.M. Seltzer: Electron and Photon Transport Programs


1. Introduction and Notes on Program DATAPAC-4. NBS 9836, 1968;
2. Notes on Program ETRAN-15. NBS 9837, 1968
(Monte Carlo Code System for Electron and Photon Transport Through
Extended Media)

ISO-PC

Revision of ISOSHLD
Engle, R.L., J. Greenborg, N.M. Hendrickson: ISOSHLD - A Computer Code
for General Purpose Isotope Shielding Analysis. BNWL-236, 1966
(X-Ray, Gamma-Ray, Bremsstrahlung Dose-Rates)

Landolt-Brnstein
New Series VIII/4

Ref. p. 5-32]

5 Shielding against ionizing radiation

5-29

Computer Codes
MCNP-4C

Briesmeister, J. F.: MCNP - A General Monte Carlo N-Particle Transport


Code. Version 4C, LA-13709-M, 2000
(Coupled Neutron, Electron, Gamma 3-D Time-Dependent Monte Carlo
Transport Calculation Code)

MORSE-CGA

Emmett, M.B.: MORSE-CGA, A Monte Carlo Radiation Transport Code


with Array Geometry Capability. ORNL-6174, 1985

NAC

Weinstein, Suzanne T.: NAC - Neutron Activation Code. NASA TM X-5260,


1968

ORIGEN2

Croff, A.G.: A Users Manual for the ORIGEN2 Computer Code.


ORNL/TM-7175, 1980

PALLAS

Takeuchi, K., S. Tanaka: PALLAS-1D(VII): A Code for Direct Integration of


Transport Equation in One-Dimensional Plane and Spherical Geometries.
JAERI-M 84-214, 1984

PENELOPE2001

Salvat, F., J.M. Fernandez-Varea, E. Acosta, J. Sempau: PENELOPE, A


Code System for Monte Carlo Simulation of Electron and Photon Transport.
Proceedings of a Workshop/Training Course, OECD/NEA 5-7 November,
2001, NEA/NSC/DOC, 2001

PUTZ

Ingersoll, D.T.: User's Manual for PUTZ - A Point-kernel Photon Shielding


Code. ORNL/TM-9803, 1986

QAD-CGGP

Sakamoto, Y., S. Tanaka: QAD-CGGP2 and G33-GP2- Revised Versions of


QAD-CGGP and G33-GP Codes with Conversion Factors from Exposure to
Ambient and Maximum Dose Equivalents. JAERI-M 90-110, 1990

RAID

Moore, J.A., J.B. Eggen, F.O. Leopard: Monte Carlo Procedure for Analysis
of Radiation in Ducts (RAID). AFWL-TR 67-9, 1967
(Gamma, Neutron Scattering in Cylindrical or Multibend Ducts)

SAM-CE

Steinberg, H.A. et al.: SAM-CE - A Monte Carlo Code for Three


Dimensional Neutron, Gamma Ray and Electron Transport (Revision 5).
MR-7052-5, 1977

SKYSHINE

Lampley, C.M., M.C. Andrew, M.B. Wells: The SKYSHINE-III Procedure:


Calculation of the Effects of a Structure Design on Neutron, Primary
Gamma-Ray and Secondary Gamma-Ray Dose Rates in Air. RRA-T8209A,
1988

SRNA-2KG

Ilic, R.D.: SRNA, Protons Transport Simulation by Monte Carlo Techniques


User's Guide. Version 2KG, 2001

Landolt-Brnstein
New Series VIII/4

5-30

5 Shielding against ionizing radiation

[Ref. p. 5-32

Table 5.6.2 Online Nuclear Data Services and Code Services


Address

Information

Atomic Mass Data Center (AMDC)


http://csnwww.in2p3.fr/AMDC/

Atomic Mass Data,


Q-Values

Gesellschaft fr Schwerionenforschung (GSI)


http://www.gsi.de/

Links to Data Banks,


Literature-Research

International Atomic Energy Agency (IAEA)


Nuclear Data Center
http://www-nds.iaea.or.at

Nuclear Structure and Decay Data,


Cross Section Data (Photons, Neutrons, charged
Particles) (ENSDF, ENDF),
Nuclear Science References

Nuclear Data Center


Japan Atomic Energy Research Institute
http://wwwndc.tokai.jaeri.go.jp/

Chart of Nuclides,
Evaluated Nuclear Data Library,
Tables of Nuclear Data

Korea Atomic Energy Research Institute (KAERI) Nuclear Structure and Decay Data,
Nuclear Data Evaluation Lab
Photon Cross Section Data
http://atom.kaeri.re.kr
Lawrence Berkeley National Laboratory (LBNL)
Isotopes Project
http://isotopes.lbl.gov

Nuclide-Table, Nuclide-Chart,
Isotope Explorer,
Internet Isotope Explorer

Los Alamos National Laboratory (LANL)


T-2 Nuclear Information Service
http://t2.lanl.gov/data/decayd.html

Nuclear Structure and Decay Data,


Cross Section Data (Photons, charged Particles,
thermal Neutrons) (ENSDF, ENDF)
Nuclide-Chart, Nuclear Data Viewer

Lunds Universitet
LUND Nuclear Data Service
http://nucleardata.nuclear.lu.se/nucleardata

Nuclear Structure and Decay Data (ENSDF),


Literature, References,
Isotope Explorer,
Internet Isotope Explorer

www-tech.mit.edu/Chemicool

Periodical System of Elements

Nuclear Energy Agency (NEA)


http://www.nea.fr/html/dbdata/

Nuclear Data, Computer Codes,


Experimental Nuclear Reaction Data Retrievals,
Evaluated Nuclear Data Retrievals,
Bibliographical Research

National Institute of Standards and Technology


(NIST)
http://physics.nist.gov/PhysRefData/contents.html

Physical Constants,
X-ray and gamma ray data,
X-Ray Attenuation and Absorption for Materials of
Dosimetric Interest,
XCOM: Photon Cross Sections Database,
Stopping-Power and Range Tables for Electrons,
Protons, and Helium Ions,
Radionuclide Half-life Measurements Made at
NIST,
Atomic Weights and Isotopic Compositions

Landolt-Brnstein
New Series VIII/4

Ref. p. 5-32]

5 Shielding against ionizing radiation

5-31

Address

Information

National Nuclear Data Center (NNDC)


http://www.nndc.bnl.gov

Nuclear Structure and Decay Data,


Neutron Cross Sections, Online-Service,
Literature, References,
Documentation of Data Banks and Computer Codes
(ENSDF, ENDF,...),
Nuclear Science References

PhysicsWeb
http://physicsweb.org/TIPTOP/paw/

Information,
Links

Radiation Safety Information Computational


Center (RSICC)
http://www-rsicc.ornl.gov/rsicc.html

Codes and Data,


Newsletter,
Workshops

Triangle Universities Nuclear Laboratory (TUNL)


http://www.tunl.duke.edu/NuclData

Nuclear Data for light Nuclides


(A = 3 to 20)

Department of Computer Science


University of Columbia (UBC)
http://www.cs.ubc.ca/elements/periodic-table

Periodical System of Elements

US Nuclear Data Program (USNDP)


http://www.nndc.bnl.gov/usndp/

Links to Nuclear Data Banks

WebElements
http://www.webelements.com/

Periodical System of Elements

Landolt-Brnstein
New Series VIII/4

5-32

5 Shielding against ionizing radiation

5.7 References
64Kno
68Gre
73Bel
73Eng
74Jae
81Mug
84ICR1
84ICR2
84Mug
88DLC
88Mcl
90Shi
91ANS
91Tan
92ICR
92Suz
93Suz
93ICR
95Hub

96ICR

Knop, G., Paul,W.: Interaction of electrons and -particles with matter; Alpha-, beta- and
gamma-Ray spectroscopy, Vol. 1, Siegbahn, K. (ed.), North-Holland Publishing Company,
1964.
Greiss, H.B.: Nukleonik 10 (1968) 283.
Bell, M.J.: ORIGEN-The ORNL isotope generation and depletion code, ORNL-4628, 1973.
Engle, jr., W.W.: A user manual for ANISN; A one dimensional discrete ordinates transport
code, ORNL-TM-4280, 1973.
Jaeger, R.G., Hbner, W.: Dosimetrie und Strahlenschutz, G. Thieme Verlag, 1974, p. 378.
Mughabghab, S.F., Divadeenam, M., Holden, N.E.: Neutron cross sections, Vol 1, Neutron
Resonance Parameters and Thermal Cross Sections, Part A, Z=1-60. New York: Academic
Press, 1981; NNDC: Online-Datenbank: http://www.nndc.bnl.gov/nndc/ensdf/ensdfindex.html
International Commission on Radiation Units and Measurements, ICRU Report 35, Radiation
dosimetry: Electron beams with energies between 1 and 50 MeV, ICRU Publications, 1984.
International Commission on Radiation Units and Measurements, ICRU Report 37, Stopping
powers for electrons and positrons, ICRU Publications, 1984.
Mughabghab, S.F.: Neutron cross sections, Vol 1, Neutron resonance parameters and thermal
cross sections, Part B, Z=61-100. New York: Academic Press, 1984; NNDC: OnlineDatenbank: http://www.nndc.bnl.gov/nndc/ensdf/ensdfindex.html
Radiation Shielding Information Center Data Package DLC-136/PHOTX, Photon Interaction
Cross Section Library, contributed by National Institute of Standards and Technology, 1988.
McLane, V., Dunford, C.L., Rose, P.F.: Neutron Cross Sections, Vol. 2. Boston: Academic
Press, 1988.
Shibata, K., Nakagawa, T., Asami, T., Fukahori, T., Narita, T., Chiba, S., Mizumoto, M.,
Hasegawa, , Kikuchi, Y., Nakajima, Y., Igarasi, S.: Japanese Evaluated Nuclear Data Library,
Version-3, -JENDL3-, JAERI1319 Japan Atomic Energy Research Institute, 1990.
American National Standard for Gamma-Ray Attenuation Coefficients and Buildup Factors for
Engineering Materials, ANSI/ANS-6.4.3-1991, 1991.
Tanaka, T., Suzuki, T.: A calculation method of photon dose equivalent based on the revised
technical standards of radiological protection, ORNL/TR-90/29, Oak Ridge National
Laboratory, 1991.
International Commission on Radiation Units and Measurements, ICRU Report 47,
Measurements of dose equivalents from external photon and electron radiations, ICRU
Publications, 1992.
Suzuki, T., Hasegawa, A., Tanaka, S., Nakashima, H.: Development of BERMUDA: A
radiation transport code system, Part I: Neutron transport codes, JAERI 1327, Japan Atomic
Energy Research Institute, 1992.
Suzuki, T., Hasegawa, A., Tanaka, S., Nakashima, H.: Development of BERMUDA: A
radiation transport code system, Part II: Gamma rays transport codes, JAERI-M 93-143, Japan
Atomic Energy Research Institute, 1993.
International Commission on Radiation Units and Measurements, ICRU Report 49, Stopping
powers and ranges for protons and alpha particles, ICRU Publications, 1993.
Hubbell, J.H., Seltzer, S.M.: Tables of X-ray mass attenuation coefficients and mass energyabsorption coefficients from 1 keV to 20 MeV for Elements Z=1 to 92 and 48 additional
substances of dosimetric interest. Online:
http://physics.nist.gov/PhysRefData/Xcom/Text/XCOM.html
International Commission on Radiological Protection, Publication 74: Conversion coefficients
for use in radiological protection against external radiation, Ann. ICRP 26, No.3/4 (1996).

Landolt-Brnstein
New Series VIII/4

5 Shielding against ionizing radiation


97ICR
01JRI
01Sak

02Nak

5-33

International Commission on Radiation Units and Measurements, ICRU Report 56, Dosimetry
of external Beta-rays for radiation protection, ICRU Publications, 1997.
The Japan Radioisotope Association, Radioisotope Pocket Data Book (Revised Edition 10),
2001 (in Japanese).
Sakamoto, Y., Endo, A., Tsuda, S., Takahashi, F., Yamaguchi, Y.: Shielding calculation
constants for use in effective dose evaluation for photons, neutrons and bremsstrahlung from
Beta-ray, JAERI-Data/Code 2000-044, Japan Atomic Energy Research Institute, 2001, (in
Japanese).
Nakagawa, T, Kawasaki, H., Shibata, K. (Eds.):Curves and tables of neutron cross sections in
JENDL-3.3, Part I (Z = 1 - 50) and Part II (Z = 51 - 100), JAERI-Data/Code 2002-020, Japan
Atomic Energy Research Institute, 2002.

Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]

6 External dosimetry

6-1

6 External dosimetry

This chapter introduces the main protection and operational quantities in external dosimetry and describes
the anthropomorphic models used for their calculations. Conversion coefficients i.e. mean organ
equivalent doses normalised to the measurable quantity air kerma free-in-air are given for idealized
geometries representing occupational exposures and for environmental source geometries.

6.1 Protection and operational quantities


6.1.1 Protection quantities
The International Commission on Radiological Protection (ICRP) for more than 50 years supports a
system for radiological protection, based on concepts, quantities, and basic recommendations. The
concept of radiation protection is based on the justification, optimisation and limitation of radiation
exposure. This concept includes a dose limitation system for occupational and man-made environmental
radiation exposures to ensure that the radiation risk would not exceed reasonable limits. The most recent
set of protection quantities recommended in ICRP60 [91ICR] includes the organ or tissue equivalent
doses HT and the effective dose E (see Chap. 4). These quantities are not measurable but can be calculated
if the exposure conditions are known. The quantity to be limited in radiation protection of occupationally
exposed persons and members of the public is the effective dose E which is the weighted mean of
equivalent doses of several organs and tissues of the body that are considered to be most sensitive.
E=

w H = w D
T

T ,R

wR

where HT is the mean organ equivalent dose and wT is the tissue weighting factor (with wT = 1) which
takes into account the differences in the stochastic radiation risk of the different organs. It is derived from
the mean organ absorbed dose DT, i.e. the total amount of energy deposited in an organ (or tissue) T per
mass of the organ, by multiplying with a radiation weighting factor wR reflecting the relative biological
effectiveness of the radiation incident on the body or emitted from radionuclides in the body. The
sensitive organs and tissues together with their respective tissue weighting factors wT were defined in
ICRP Publication 60 [91ICR] (see Chap. 4).

6.1.2 Operational Quantities


The International Commission on Radiation Units and Measurements (ICRU) has defined a set of
operational quantities for area and individual monitoring [85ICR, 92ICR1, 93ICR] in response to the
recommendations of the International Commission on Radiological Protection [77ICR] which were
Landolt-Brnstein
New Series VIII/4

6-2

6 External dosimetry

[Ref. p. 6-42

designed to provide an estimate of the protection quantities defined by ICRP and to serve as calibration
quantities for dosimeters used in monitoring. For area monitoring, the appropriate operational quantities
are the ambient dose equivalent H*(d), and the directional dose equivalent, H'(d,), both defined at a
depth d, on the principal axis of the 30 cm diameter ICRU sphere. The recommended value of d for
strongly penetrating radiation is 10 mm and for weakly penetrating radiation it is 0.07 mm (see Sect.
4.5.3.3).
For individual monitoring, the quantity personal dose equivalent Hp(d) was proposed, which is the
dose equivalent in soft tissue, at an appropriate depth d below a specified point on the body [92ICR1,
93ICR]. For weakly penetrating radiation, depths of 0.07 mm for the skin and 3 mm for the eye lens are
used, denoted by Hp(0.07) and Hp(3), respectively; for strongly penetrating radiation, a depth of 10 mm is
currently recommended by the ICRU, denoted by Hp(10) (see Sect. 4.5.3.4).
Personal dose equivalent is defined in the human body and may, therefore, vary between individuals;
furthermore, the depth d is specified but the position of the point below which it is defined is not fixed but
only correlated to the position of the dosemeter worn on the body. Consequently, the personal dose
equivalent can be expected to vary also between locations on any given individual and is, hence,
anticipated to be a multi-valued quantity [96ICR, 98ICR, 99Zan]. To make this quantity single-valued in
a given exposure situation, both a particular location on the human body and a particular phantom of the
body need to be specified for evaluation.
For calibration purposes, surrogate quantities for Hp(d) have been introduced: it is recommended
that personal dosimeters normally worn on the trunk are calibrated on an ICRU tissue slab or PMMA
(polymethylmethacrylate) slab with dimensions 30 30 15 cm3 [92ICR1]. Conversion coefficients for
personal dose equivalent at the relevant depths d in the ICRU tissue slab Hp,slab(d) have been calculated
for calibration purposes [91Gro, 95ISO, 95Til] and have been recommended for use [96ICR, 98ICR,
98Cla].
The operational quantities used in measurement were designed to provide a reasonable estimate of the
appropriate protection quantity. For external exposures of the body in a given field, it is desirable that the
ratio of the value of the appropriate protection quantity to the value of the corresponding operational
quantity is less than unity, i.e. the operational quantity should always provide a conservative estimate of
the protection quantity.
More about the definitions of the operational quantities can be found in Chap. 4.

6.2 Dosimetric models


6.2.1 Models and phantoms of the human body
To estimate the protection quantities organ and tissue equivalent doses HT there are two approaches,
an experimental and a theoretical one. The experimental determination is very difficult whereas the
mathematical modelling of an exposure has been proved to be extremely flexible and powerful. For
this purpose, a series of computer models of the human body were designed in the past, together with
computer codes simulating the radiation transport and energy deposition in the body.
The computer models used for the representation of the human body in dose calculations can range
from simple geometric forms such as spheres, cylinders or slabs to complex representations of detailed
anatomical features. Such complex models, used since 1966 for the estimation of organ doses are the socalled mathematical phantoms, which are models whose body organs and tissues are described by
mathematical expressions representing planes or cylindrical, conical, elliptical or spherical surfaces. The
mostly used model was the MIRD one, named after the initials of the Medical Internal Radiation Dose
Committee of the US Society of Nuclear Medicine where it was initially developed [69Sny, 78Sny]. From
this, several paediatric models were derived to represent infants and children of various ages, for example
those from Cristy [80Cri]. As an improvement to these hermaphrodite models, separate male and female

Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]

6 External dosimetry

6-3

adult mathematical models have been introduced by Kramer et al. [82Kra] called Adam and Eva. For
these models, the organ masses and volumes are in accordance with the ICRP data on Reference Man
[75ICR]; in addition to a separation of the gender-specific organs, the phantom Eva is smaller than Adam,
according to the difference in size of the male and female Reference Man. The oesophagus, an organ
which had not originally been defined in these models, was incorporated in the form of an elliptical
cylinder ranging in height from within the neck down to the top of the stomach and lying in front of the
spine, slightly shifted to the left side [92Zan].
Adam and Eva contain all organs and tissues relevant for the evaluation of effective dose, with only
few exceptions: since there is no specific representation of the bone surface, the skeleton is modelled as
a homogeneous mixture of all skeletal constituents, i.e., hard bone, bone marrow and certain peri-articular
tissues. Commonly, the dose to this representation of the entire skeleton is taken to represent the dose to
the bone surface. Although there may be certain differences, these are usually considered to be small in
view of the small weighting factor (wT=0.01) assigned to this tissue. The muscles were represented by
that part of the body volume not attributed to any other organ or tissue of the models.
More recently, four models representing the adult female, non-pregnant and at 3 stages of pregnancy
were elaborated by Stabin et al [99Sta]. A comprehensive review of models and phantoms of the human
body can be found in ICRU Report 48 [92ICR2]. The term model refers to computational models,
whereas phantom implies either a physical phantom or a computational one. Spherical and slab
phantoms are convenient and simple approximations of the human body.
A spherical model of 30 cm diameter made of ICRU tissue-equivalent material (see Sect. 4.5.3.3) is
used for the definition of the operational quantities. Various tissue substitutes are available for fabrication
of corresponding physical phantoms, including tissue-equivalent material, water and perspex. For
calibration purposes, slab tissue-substitute phantoms of 30 30 15 cm3 are used.
Recently a new generation of computational phantoms has become available which offer the prospect
of increased realism and accuracy in dose calculations. These models use computed (CT) or magnetic
resonance (MR) tomographic data of real persons to provide three-dimensional representations of the
human body and comprise a large number of volume elements (voxels) all of the same size but with
differing composition according to the organ to which they belong. The GSF-National Research Centre in
Germany started since the mid eighties the development of voxel models covering various ages and
anatomical statures [88Zan, 01Zan, 02Pet]. Due to their anatomical realism, such models have been the
subject of increasing interest and acceptance, and others have been developed also elsewhere [94Zub,
96Dim, 00Xu].
Both MIRD-type and voxel models incorporate different densities and atomic compositions for the
various body tissues. The number of organs simulated varies from model to model, however, the latest
versions include all organs defined to be important. Fig. 1 shows views of selected organs of the
mathematical model Eva [82Kra] and the adult female voxel model Donna, developed at GSF [02Pet].

Landolt-Brnstein
New Series VIII/4

6-4

6 External dosimetry

[Ref. p. 6-42

Fig. 6.1. View of selected organs of the mathematical


model Eva [82Kra] and the adult female voxel model
Donna, developed at GSF [02Pet].

6.2.2 Idealized geometries representing occupational exposures


To simulate occupational exposure conditions, whole-body irradiation with idealised geometries are
conventionally taken into account. These include broad parallel beams and fully isotropic radiation
incidence. The directions of incidence for the parallel beams considered are: anterior-posterior (AP),
posterior-anterior (PA), left lateral (LLAT), right lateral (RLAT) and a full 360 rotation around the
phantoms longitudinal axis (ROT). Although these geometries are idealised, they may be taken as
acceptable approximations to actual conditions of exposure. The AP, PA and both lateral geometries are
supposed to approximate radiation fields from single sources and particular body orientations. The ROT
geometry approximates the exposure of a person who moves randomly in the field of a single source
irradiating at right angles to the longitudinal axis of the body. The fully isotropic (ISO) source simulates
the geometry of a body suspended in a large cloud of radioactive gas.

6. 2. 3 Environmental source geometries


For external exposures to environmental sources the dosimetric quantities of interest are the radiation
doses received by the radiosensitive organs and tissues of the body due to photons and electrons emitted
by radionuclides distributed in soil and air. The radiation dose depends strongly on the temporal and
spatial distribution of the radionuclide to which a human is exposed. The situation of radioactive release
in water is more rare and is not covered here. The kinds of radiation of concern are those sufficiently
Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]

6 External dosimetry

6-5

penetrating to traverse the overlying tissues of the body and which deposit their energy in organs and
tissues of the body. Penetrating radiations are limited to photons and electrons. Neutrons from cosmic
radiation are not dealt here.
For simulating the exposure to environmental gamma-rays, the following three typical cases of
environmental sources are considered here: (1) semi-infinite volume source in the air; (2) infinite plane
source in the ground; (3) semi-infinite volume source in the ground. The first source configuration models
the gaseous radioactive release into the atmosphere at locations which are not too near to the release
point, by assuming a homogeneous contamination of the air up to a height of 1000 m above a smooth airground interface. The second source simulates the deposition of radionuclides in the ground, by assuming
an infinite plane source in the soil. The third source simulates the natural radioactivity in the ground (the
dominant radionuclides of the 238U series, the 232Th series and 40K) being homogeneously distributed to a
depth of 1 m in the soil.

6.2.4 Methods of calculating protection quantities in computational models


Today the predominant method for assessment of absorbed doses in the body is the application of Monte
Carlo methods to simulate the transport of radiation in the body. The organ and tissue doses are then
estimated in the form of conversion coefficients giving organ doses per unit of a measurable quantity.
The Monte Carlo method is a computational model in which physical quantities are calculated by
simulating the transport of particles. In the computer program, single particles are followed through their
histories of inelastic and elastic scattering or absorption within the anthropomorphic model. Depending
on their energy and on the material they are passing through, the particles interact differently and each
mode of interaction has a certain probability of occurring, which can be selected by appropriate use of
random numbers and probability distributions. Individual particles have different energies, directions and
path lengths modelled randomly from probability distributions. By averaging over large numbers of
random paths, good estimates of the quantities of interest can be made. Basic elements of Monte Carlo
simulation include the choice of random number generator which provides the method of sampling the
cross-section data and coordinate transformations from probability distributions. The mean absorbed dose
in a defined volume of material is computed from the incident and emerging energies from the volume by
dividing the energy imparted by the mass of the volume material.
Most codes dealing mainly with photon interaction assume that electrons generated through different
interactions are absorbed on the spot. The energy transferred at a point of inelastic photon interaction is
then modelled as being deposited at that point, without considering the energy transport by secondary
charged particles (kerma approximation). This approach is valid as long as there is approximate
secondary charged particle equilibrium, which can be supposed in most cases due to the macroscopic
approach considering mean organ and tissue doses. This is acceptable for photon energies up to 3 MeV.
For the skeleton, however, the boundary effects do have an impact on the tissue dose and corrections for
secondary electron effects in the skeleton have to be applied. For neutrons up to 20 MeV the kerma
approximation introduces no significant error due to the short range of the recoil protons and heavier
charged particles.
For the estimation of absorbed doses distributions in the body, several transport codes are used and
their description is beyond the scope of this book. General Monte Carlo codes are available, such as
EGS4 [85Nel, 94Hir], ITS [92Hal] and MCNP4 [91Bri]. Various other research institutes have also
developed their Monte Carlo codes like the GSF-National Research Center code [82Kra, 89Vei], PTB/PG
[86Gro, 94Gro], etc.
The anthropomorphic models used for the collection of the data shown below were the mathematical
Adam and Eva and the mathematical MIRD hermaphrodite for photons and neutrons; for electrons, the
ICRU tissue sphere and slab as well as Adam and Eva were employed.

Landolt-Brnstein
New Series VIII/4

6-6

6 External dosimetry

[Ref. p. 6-42

A joint task Group of ICRP and ICRU reviewed the conversion coefficients reported by different
researchers. Conversion coefficients for external photon, neutron and electron exposures and the idealised
geometries AP, PA, LLAT, RLAT and ROT were evaluated and compiled to be used as reference data;
they can be found at the ICRP Report 74 [96ICR] and ICRU Report 57 [98ICR].

6.3 Conversion coefficients for photons


6.3.1 Occupational
This Section provides organ doses in the form of conversion coefficients, i.e. mean organ equivalent
doses normalised to air kerma free-in-air which is a measurable quantity; the conversion coefficients
are given in the unit SvGy1. No location for a measurement of the normalisation quantity has to be
specified since for the parallel and ideally isotropic geometries the photon fluence is invariant throughout
the field.
The organ equivalent dose conversion coefficients were calculated for the male Adam and female Eva
models [82Kra] separately. Average organ equivalent dose conversion coefficients were computed as the
arithmetic mean of those for the male and female models. The gonad equivalent dose conversion
coefficients are the arithmetic mean values of the respective coefficients for testes and ovaries.
For these calculations, the GSF Monte Carlo code was used which computes the dose deposited by
photons from an external or internal source in various sections of a different media model of the body.
The code is based on the fractional photon technique and uses the kerma approximation. The latter is
valid only when charged particle equilibrium is established which can be supposed in most cases due to
the moderate differences of the photon cross sections for the tissues in the human body and the
macroscopic approach of energy deposition. There are two exceptions where boundary effects do have an
impact on the tissue dose. One is the red bone marrow, where a moderate increase in energy deposited in
the marrow cavities is expected from increased photoelectron emission from the surrounding bone. For
photon exposures this effect was accounted for by applying appropriate correction factors [69Spi, 97Zan]
to the energy deposited to the red bone marrow calculated using the kerma approximation. The other
tissue where boundary effects could be of consequence is the bone surface, a very thin soft tissue layer
enveloping the bones. Here secondary electron equilibrium is not valid for energies below approximately
300 keV as there the bone cross section values are considerably higher than those for soft tissues,
resulting in an increased production of secondary electrons in the bones and, consequently, a dose
enhancement at the interface between the bones and the adjacent soft tissues compared to the dose to
tissue beyond the range of these secondary electrons. This enhanced dose to the tissue adjacent to bones
is, however, not as high as the mean dose to the homogeneous mixture of skeletal tissues [68Dre].
Consequently this can be taken as a conservative estimate of the dose to the bone surface in this photon
energy range. Above 300 keV, the cross sections of bone and soft tissue per mass density have a similar
magnitude, and approximate secondary electron equilibrium is established.
To the calculated values of the conversion coefficients for monoenergetic photons, a fitting procedure
using cubic spline functions was applied. With these fitting functions, values were also evaluated for 200
photon energies distributed equidistantly on a logarithmic scale between 10 or 15 keV and 10 MeV.
Figures 6.2-6.8 show the conversion coefficients as a function of photon energy for 8 selected organs and
tissues and for AP, PA, LLAT, RLAT, LAT, ROT and ISO geometries, respectively. These are the
average values evaluated as the arithmetic mean of those for the male (Adam) and female (Eva) model.
The complete sets of organ equivalent doses for the male, female and average can be found in Zankl et.
al. [97Zan] shown graphically as well as in tabular form. The average values of the male and female
models denoted as adult are also adopted as reference values for conversion coefficients and are
presented in detail in ICRP Report 74 [96ICR] and ICRU Report 57 [98ICR] for those specific organs for
which the ICRP recommends tissue weighting factors (see Chap. 4, Table 3).

Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]

6 External dosimetry

6-7

Organ equivalent dose per unit air kerma [Sv/Gy]

The energy dependence of the conversion coefficients for single organs is determined by the photon
interaction cross sections in tissues, the location of the organ in the body and the irradiation geometry.
The cross sections decrease with increasing photon energy and the conversion coefficients
correspondingly increase due to the increasing range of photons in the body. With further increasing
energy and range of the photons, the conversion coefficients decrease. This leads to more or less
pronounced peak in the energy range between 80-100 keV. The more pronounced peak of the conversion
coefficients of skeleton is due to the large values of the ratio of the attenuation coefficients of bone and
air respectively.
Table 6.1 shows the effective dose per unit air kerma as a function of energy for the six irradiation
geometries, calculated for adults using the models Adam and Eva. The different forms of the energy
dependence of the conversion coefficients for effective dose with irradiation geometry result from the
different locations of the organs relative to the incoming photon beam and the value of their tissue
weighting factors. As it can be seen from Table 6.1, the conversion coefficients of E for AP irradiation are
always higher than the corresponding ones for other irradiation geometries. This is due to the fact that for
AP photon incidence most organs with large tissue weighting factors are anteriorly located.

gonads

Parallel AP

lungs
RBM
stomach
liver
thyroid
skin
skeleton

0
0.01

0.1

Photon energy [MeV]

10

Fig. 6.2. Organ equivalent doses per unit air kerma free-in-air for selected organs in AP irradiation as a function of
photon energy, evaluated as the arithmetic mean of those for the male (Adam) and female (Eva) model;[97Zan].

Landolt-Brnstein
New Series VIII/4

6-8

6 External dosimetry

[Ref. p. 6-42

Organ equivalent dose per unit air kerma [Sv/Gy]

3
gonads

Parallel PA

lungs
RBM
stomach
liver
thyroid
skin
skeleton

0
0.01

0.1

Photon energy [MeV]

10

Fig. 6.3. Organ equivalent doses per unit air kerma free-in-air for selected organs in PA irradiation as a function of
photon energy, evaluated as the arithmetic mean of those for the male (Adam) and female (Eva) model; [97Zan].

Organ equivalent dose per unit air kerma [Sv/Gy]

2
gonads
lungs
RMB
stomach
liver
thyroid
skin
skeleton

Parallel LLAT

0
0.01

0.1

Photon energy [MeV]

10

Fig. 6.4. Organ equivalent doses per unit air kerma free-in-air for selected organs in LLAT irradiation as a function
of photon energy, evaluated as the arithmetic mean of those for the male (Adam) and female (Eva) model; [97Zan].

Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]

6 External dosimetry

6-9

Organ equivalent dose per unit air kerma [Sv/Gy]

2
gonads

lungs
RBM
stomach
liver
thyroid
skin

Parallel RLAT

skeleton

0
0.01

0.1

Photon energy [MeV]

10

Fig. 6.5. Organ equivalent doses per unit air kerma free-in-air for selected organs in RLAT irradiation as a function
of photon energy, evaluated as the arithmetic mean of those for the male (Adam) and female (Eva) model; [97Zan].

Organ equivalent dose per unit air kerma [Sv/Gy]

2
gonads

lungs
RBM
stomach
liver
thyroid
skin

Parallel LAT

skeleton

0
0.01

0.1

Photon energy [MeV]

10

Fig. 6.6. Organ equivalent doses per unit air kerma free-in-air for selected organs in LAT irradiation as a function of
photon energy, evaluated as the arithmetic mean of those for the male (Adam) and female (Eva) model; [97Zan].

Landolt-Brnstein
New Series VIII/4

Organ equivalent dose per unit air kerma [Sv/Gy]

6-10

6 External dosimetry

[Ref. p. 6-42

gonads

Parallel ROT

lungs
RBM
stomach
liver
thyroid
skin

skeleton

0
0.01

0.1

Photon energy [MeV]

10

Organ equivalent dose per unit air kerma [Sv/Gy]

Fig. 6.7. Organ equivalent doses per unit air kerma free-in-air for selected organs in ROT irradiation as a function of
photon energy, evaluated as the arithmetic mean of those for the male (Adam) and female (Eva) model; [97Zan].

gonads

Parallel ISO

lungs
RBM
stomach
liver
thyroid
skin

skeleton

0
0.01

0.1

Photon energy [MeV]

10

Fig. 6.8. Organ equivalent doses per unit air kerma free-in-air for selected organs in ISO irradiation as a function of
photon energy, evaluated as the arithmetic mean of those for the male (Adam) and female (Eva) model; [97Zan].

Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]

6 External dosimetry

6-11

Table 6.1. Effective dose E per unit air kerma free-in-air Ka for monoenergetic photons and various
irradiation geometries. Data are from Zankl et. al. [97Zan], calculated using the male (Adam) and female
(Eva) model.
Photon
energy
[MeV]
0.010
0.015
0.020
0.030
0.040
0.050
0.060
0.070
0.080
0.100
0.150
0.200
0.300
0.400
0.500
0.600
0.800
1.000
2.000
4.000
6.000
8.000
10.000

E/Ka
[SvGy1]
AP
0.00654
0.0402
0.122
0.416
0.787
1.104
1.306
1.405
1.431
1.392
1.255
1.172
1.091
1.055
1.035
1.024
1.010
1.002
0.992
0.992
0.993
0.991
0.990

PA
0.00248
0.00590
0.0183
0.129
0.372
0.641
0.847
0.968
1.020
1.031
0.960
0.916
0.881
0.872
0.870
0.871
0.875
0.881
0.901
0.918
0.924
0.928
0.929

LLAT
0.00173
0.00550
0.0156
0.0907
0.242
0.406
0.529
0.599
0.630
0.643
0.622
0.616
0.616
0.624
0.636
0.648
0.671
0.692
0.758
0.813
0.836
0.850
0.860

RLAT
0.00172
0.00551
0.0151
0.0782
0.204
0.344
0.454
0.521
0.553
0.570
0.551
0.548
0.556
0.570
0.585
0.600
0.627
0.651
0.728
0.796
0.827
0.846
0.860

ROT
0.00326
0.0154
0.0463
0.191
0.427
0.661
0.828
0.924
0.961
0.960
0.893
0.854
0.824
0.814
0.812
0.814
0.821
0.831
0.871
0.909
0.925
0.934
0.941

ISO
0.00271
0.0123
0.0362
0.144
0.326
0.511
0.642
0.720
0.749
0.748
0.700
0.679
0.664
0.667
0.675
0.685
0.703
0.719
0.774
0.824
0.846
0.859
0.868

Figures 6.8 and 6.9 demonstrate the age dependence for effective dose, by showing the effective dose
for adults as well as for 0-, 1-, 5-, 10-, and 15-year old children for AP and ROT geometry respectively.
The data of this figure stem from Yamagushi [94Yam] who used hermaphrodite paediatric and adult
mathematical models of different body sizes developed by Cristy [80Cri]. It can be seen that the smaller
body size results in higher organ dose conversion coefficients, and consequently higher effective doses,
particularly at low photon energies. The largest variation of effective dose with age was found for the
LAT and ISO geometries. Similarly, Zankl et. al. [97Zan] have shown that the conversion coefficients for
the female model Eva are approximately 2 % to 20 % higher than those for the male model, depending on
photon energy, due to the slightly smaller body size of the female model. For AP irradiation, the lung
dose conversion coefficients for the female model are between 5 % and 20 % lower than those for the
male, as for this geometry the lungs of the female model are partially shielded by the breast. Furthermore,
some differences were observed for the organ conversion coefficients calculated by different authors and
are mainly due to the different human models used in the calculations and occur at low photon energies:
the female model Eva, used for the calculations of Zankl et. al. has a smaller body size than the
hermaphrodite model used by Yamaguchi, resulting in higher organ conversion coefficients particularly
for low energies. Consequently, differences (up to 20 %) between the effective dose coefficients from
adults were observed. For energies above 70 keV there was general agreement within the statistical
uncertainties.

Landolt-Brnstein
New Series VIII/4

Effective dose per unit air kerma E/Ka [Sv/Gy]

6-12

6 External dosimetry

[Ref. p. 6-42

Parallel AP

0 year
1 year
5 years
10 years
15 years
adult

0
0.01

0.1

Photon energy [MeV]

10

Fig. 6.9. Effective dose per unit air kerma free-in-air for AP irradiation geometry calculated for MIRD-type
hermaphrodite phantoms of various ages; [94Yam].

Effective dose per unit air kerma [Sv/Gy]

Parallel ROT
1

0 year
1 year
5 years
10 years
15 years
adult

0
0.01

0.1

Photon energy [MeV]

10

Fig. 6.10. Effective dose per unit air kerma free-in-air for ROT irradiation geometry calculated for MIRD-type
hermaphrodite phantoms of various ages; [94Yam].

Conversion coefficients for the operational quantities ambient dose equivalent and directional dose
equivalent are shown in Table 6.2. These are values recommended by the ICRP [92ICR1] and stem from
calculations by several groups using Monte Carlo methods on the ICRU sphere assuming electronic
equilibrium.
Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]

6 External dosimetry

6-13

Table 6.2. Conversion coefficients for air kerma free-in-air Ka, directional dose equivalent H(0.07,0),
and ambient dose equivalent H*(10), per unit fluence of monoenergetic photons; [92ICR1].
Photon energy
Ka/
H(0.07,0)/
H* (10)/
[MeV]
[pSv cm2]
[pGy cm2]
[pSv cm2]
0.010
7.60
7.20
0.061
0.015
3.21
3.19
0.83
0.020
1.73
1.81
1.05
0.030
0.739
0.90
0.81
0.040
0.438
0.62
0.64
0.050
0.328
0.50
0.55
a
0.060
0.292
0.51
0.080
0.308
0.53
0.100
0.372
0.61
0.150
0.600
0.89
0.200
0.856
1.20
0.300
1.38
1.80
0.400
1.89
2.38
0.500
2.38
2.93
0.600
2.84
3.44
0.800
3.69
4.38
1.0
4.47
5.20
1.5
6.12
6.90
2.0
7.51
8.60
3.0
9.89
11.1
4.0
12.0
13.4
5.0
13.9
15.5
6.0
15.8
17.6
8.0
19.5
21.6
10.0
23.2
25.6
a

H(0.07,0) is not accurately determined at energies above 60 keV since there is no electronic
equilibrium.
Comparing tables 6.1 and 6.2, it can be seen that for photons with energies up to 10 MeV and
irradiation geometries AP, PA and ROT, the operational quantity H*(10) always overestimates the
protection quantity E, i.e. E/H*(10) < 1.

6.3.2 Conversion coefficients for environmental gamma ray fields


6.3.2.1 Calculation of doses for monoenergetic photons
As already mentioned above, for simulating the exposure to environmental gamma-rays, the following
three typical cases of environmental sources are considered to be representative: (a) semi-infinite volume
source in the air; (b) infinite plane source in the ground; (c) semi-infinite volume source in the ground.
The first source configuration models the gaseous radioactive release into the atmosphere at locations
which are not too near to the release point, by assuming a homogeneous contamination of the air up to a
height of 1000 m above a smooth air-ground interface. The second source simulates the deposition of
radionuclides in the ground, by assuming an infinite plane source in the soil. The source is shielded by a
soil slab of 0.5 g cm2, allowing for some surface roughness and initial migration into soil with
precipitation. The third source simulates the natural radioactivity in the ground (e.g. radionuclides of the
238
U series, the 232Th series and 40K) being homogeneously distributed to a depth of 1 m in the soil.
Landolt-Brnstein
New Series VIII/4

6-14

6 External dosimetry

[Ref. p. 6-42

In source (a), the dominant gamma rays come almost isotropically from the upper 2 directions, while
only a small amount of scattered gamma-rays comes from the lower 2 directions. Source (c) shows the
inverse tendency: the angular distribution is nearly uniform in the lower 2 directions with small
components of scattered gamma-rays stemming from the upper 2 directions. In source (b), quite a large
portion of the gamma-rays comes from horizontal directions. When the source distribution in the
environment varies from the three typical source distributions, the angular and energy distributions also
change.
To estimate the organ doses from environmental photon sources presented in this book, a three-step
procedure [91Pet] was followed:
(1) Calculation of the gamma-ray transport in the environment (monoenergetic gamma-rays and natural
radionuclides);
(2) Simulation of a secondary source around the phantom;
(3) Calculation of organ doses due to the secondary sources.
The result of this procedure, is a set of dose conversion coefficients for monoenergetic photons. Using
those, and considering the energies and intensities of the radiations emitted during nuclear
transformations of these nuclides, conversion coefficients for specific radionuclides can be computed to
relate a measurable quantity i.e. activity concentration or air kerma to the non-measurable quantities of
organ dose.
The photon transport in the environment was simulated with the Monte Carlo code YURI [85Sai], a
code specially developed for environmental problems. Compton scattering, photoelectric absorption and
pair production were considered as photon interaction processes. Air and ground were assumed to contact
each other with an infinite plane. The cross Sections used were from Storm and Israel [70Sto]. Air was
assumed to have a constant density of 1.2103 g cm3, corresponding to a temperature of 20 C and an air
pressure of 0.1 MPa and to consist of N2, O2 and Ar having weight fractions of 75.5 %, 23.2 % and 1.3 %,
respectively. Soil was taken to consist of SiO2, Al2O3, Fe2O3 and H2O with weight fractions of 58.3 %,
16.7 %, 8.3 % and 16.7 %, respectively. A soil density of 1 g cm3 has been assumed in the calculations,
since this value represents reasonably well the upper 2 cm of soil. It should be noted that the
environmental transport calculations were performed without the presence of the phantom; however, the
perturbation caused by the human body was investigated and found to be insignificant.
From the transport calculations in the environment, double differential fluences, currents (i.e. fluences
multiplied by the cosine of the angle of incidence) and air kerma values are obtained for points from 0 to
2 m above ground in steps of 20 cm. Table 6.3 shows calculated values of the air kerma rate free-in-air at
1 m height above the ground per unit activity concentration for a semi-infinite volume source in air and
per unit activity per area for an infinite plane source in the ground; Table 6.4 shows the air kerma free-inair at 1 m height above the ground per disintegration/kg for the semi-infinite volume source in the ground
due to the natural radionuclides.
Table 6.3. next page
Table 6.4. Calculated air kerma at 1 m height above the ground per disintegration/kg for a semi-infinite
volume source in the ground due to natural radionuclides; [95Sai].
Radionuclides
Air kerma per unit source intensity
[Gy / (disintegration/kg)]
238
U series
1.291013
232
Th series
1.681013
40
K
1.161014

Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]

6 External dosimetry

6-15

Table 6.3. Calculated air kerma rate at height 1 m above the ground per unit of activity concentration for
a semi-infinite volume source in air and per unit of the activity per unit area for an infinite plane source in
the ground; [95Sai].
Volume source in air
Plane source in ground
Energy
Air kerma rate per unit
Air kerma rate per unit
[MeV]
of activity concentration of activity per unit area
[(Gy s-1)/ (Bq m-3)]
[(Gy s-1)/ (Bq m-2)]
15
0.015
1.4710
8.061019
15
0.020
7.721018
1.7110
15
0.030
2.1210
2.631017
15
0.040
2.4010
3.591017
15
0.050
2.8110
4.141017
15
0.060
3.3110
4.651017
15
0.070
3.7910
5.351017
0.080
4.361015
5.981017
15
0.100
5.5510
7.541017
15
0.150
8.6810
1.211016
14
0.200
1.2010
1.681016
14
0.300
1.8710
2.611016
14
0.500
3.2110
4.341016
14
0.700
4.5610
5.901016
14
1.000
6.5810
8.091016
14
1.500
9.0810
1.131015
13
2.000
1.3210
1.411015
13
3.000
1.9610
1.911015
13
6.000
3.8510
3.191015
13
10.000
6.2610
4.811015
These height-dependent double differential (with respect to angle of incidence and photon energy)
gamma ray fields were then incorporated into the organ dose calculation with anthropomorphic models,
by establishing a secondary cylindrical source around the model to simulate the gamma-ray fields after
the results of the transport calculation in the environment (step 2 of the procedure mentioned above)
[91Pet]. The anthropomorphical models are standing on the soil modelled as a planar air/ground interface.
Scatter and absorption of the radiation in both air and ground was considered in the calculation. The
Monte Carlo code used for the transport calculation in the body was the GSF code mentioned above. The
interactions considered were photoelectric absorption, Compton scattering and pair production and the
cross section data were taken from ORNL [83Rou]. Dose conversion coefficients for Adam and Eva (see
previous Section) and several organs, including the critical ones, were estimated and can be found in
Zankl et. al. [97Zan]. From the dose conversion coefficients of the sex-specific models Adam and Eva,
conversion coefficients for an adult were derived as arithmetic average.
In Fig. 6.10 dose conversion coefficients are given for some selected organs for submersion in a
radioactive cloud (volume source in air) and in Fig. 6.11 for surface contamination. The conversion
coefficients are for an adult and are expressed as equivalent doses normalised to air kerma free-in-air at
height 1 m above the ground in Sv Gy1 as a function of photon energy. For the volume source in the
ground, conversion coefficients for the photon energy distributions corresponding to the natural
radionuclides of the decay series of 238U, 232Th and 40K are tabulated in Table 6.5.

Landolt-Brnstein
New Series VIII/4

6-16

6 External dosimetry

[Ref. p. 6-42

Table 6.5. Organ equivalent dose conversion coefficients for the natural radionuclides for an adult,
evaluated as the arithmetic mean of those for the male (Adam) and female (Eva) model; [97Zan].
Organ

Adrenals
Bladder
Brain
Colon
Eye lenses
Gonads
Kidneys
Liver
Lungs
Muscle
Oesophagus
Pancreas
Red bone marrow
Skeleton
Skin
Small intestine
Spleen
Stomach
Thymus
Thyroid
Effective dose

Organ equivalent dose per unit air kerma free-in-air at


1 m above ground [Sv Gy1]
238
232
40
U series
Th series
K
0.589
0.617
0.634
0.648
0.681
0.692
0.689
0.715
0.727
0.627
0.655
0.659
0.872
0.876
0.947
0.682
0.681
0.738
0.674
0.700
0.700
0.658
0.684
0.692
0.709
0.732
0.740
0.737
0.761
0.767
0.608
0.635
0.638
0.600
0.627
0.662
0.656
0.680
0.687
0.770
0.792
0.764
0.849
0.863
0.861
0.620
0.651
0.652
0.646
0.699
0.703
0.660
0.671
0.684
0.675
0.753
0.733
0.659
0.776
0.731
0.672
0.695
0.709

For the environmental irradiation geometries, the dependence of the organ equivalent dose conversion
coefficients on photon energy is much more uniform than for the unidirectional geometries considered for
occupational radiation exposures, and depends less on the position of the organ in the body. As the
radiation comes from all directions, every organ is quasi deep-lying relative to at least a considerable part
of the incoming photons.
The conversion coefficients for the female model were found to be up to 5 % higher than those for the
male model, due to the slightly smaller body size of the female model. Considering the two different
source types, it can be seen that the equivalent dose conversion coefficients for the volume source in air
are generally lower than those for the plane source in the ground. This results from the different angular
distribution of the radiation impinging on the body: the gamma-ray field from a source in the air is nearly
isotropic with respect to directions from the upper hemisphere, while the incident directions of the gamma
rays from a plane source have strong horizontal bias, and most photons come from horizontal directions.
Since the human body standing vertically has a reduced shielding effect for photons coming from
horizontal directions, this leads to the higher doses resulting from this geometry. However, in most cases,
the differences in the conversion coefficients were found to be less than 20 %.
Saito et. al. [98Sai] investigated the variation of effective dose for environmental gamma-rays for
source distributions other than these three typical ones and for a lying posture further to the standing one.
The change of posture of a human body and the biases of environmental sources were found to affect the
effective dose by some tens percent. A similar trend is anticipated for the individual organ doses.
Therefore, it could be concluded that the conversion coefficients for the three typical environmental
sources can be used as a reference set of values to derive the organ doses and effective doses of adults
from air kerma or source activity obtained by measurement for a variety of environmental source
configurations.

Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]

6 External dosimetry

6-17

Organ equivalent dose per unit air kerma [Sv/Gy]

2
gonads

lungs
RBM
stomach
liver
thyroid
skin

Volume source in air

skeleton
effective dose

0
0.01

0.1

Photon energy [MeV]

10

Fig. 6.11. Organ equivalent doses per unit air kerma at 1 m above the ground for some selected organs of an adult for
a volume source in air, evaluated as the arithmetic mean of those for the male (Adam) and female (Eva) model;
[97Zan].

Organ equivalent dose per unit air kerma [Sv/Gy]

gonads

lungs
RBM
stomach
liver
thyroid
skin

Plane source in ground

skeleton
effective dose

0
0.01

0.1

Photon energy [MeV]

10

Fig. 6.12. Organ equivalent doses per unit air kerma at 1 m above the ground for some selected organs of an adult for
plane source in ground, evaluated as the arithmetic mean of those for the male (Adam) and female (Eva) model;
[97Zan].

Landolt-Brnstein
New Series VIII/4

6-18

6 External dosimetry

[Ref. p. 6-42

6.3.2.2 Calculation of doses for radionuclides


Kerma rates in air and equivalent dose rates in organs for radionuclides are obtained from dose
conversion coefficients for the monoenergetic sources hT,i by multiplication with the yield yiN (in number
of photons Bq1) of photons with energy i per disintegration and summing over the photon energies of the
emission spectrum of radionuclide N:
gN =

N
i

hT,i

Kerma rates in air calculated with Monte Carlo methods in the environment due to monoenergetic
photon sources distributed exponentially in the soil or homogeneously in the air are given in Tables 6.3
and 6.4 respectively. Radionuclide-specific results are given in Table 6.6.
Table 6.6. Kerma-rates in air at 1 m above ground per unit activity per unit area (nGy h-1 per kBq m-2)
and per activity concentration in air; [94ICR].
Radionuclide Source in soil Volume
Radionuclide Source in soil Volume
source in air
source in air
at a depth of
at a depth of
0.5 g cm2
0.5 g cm2
Kerma rate
Kerma rate
Kerma rate
Kerma rate
1
1
1
[(nGy h )/
[(nGy h1)/
[(nGy h )/
[(nGy h )/
3
(kBq m )]
(kBq m3)]
(kBq m2)]
(kBq m2)]
Be-7
Nb-93m
1.67 101
1.29 102
1.10 102
6.19 104
0
1
0
Na-22
Nb-95
6.97 10
2.49 10
5.11 10
1.80 101
1
1
1
Na-24
Nb-95m
1.10 10
2.40 10
9.72 10
1.59 102
1
2
0
K-40
Nb-97
4.62 10
2.21 10
3.78 10
1.52 101
1
2
2
K-42
Mo-93
8.53 10
7.58 10
6.52 10
3.54 103
0
1
1
Sc-46
Mo-99
6.31 10
4.85 10
4.75 10
3.43 102
1
3
1
Cr-51
Tc-99m
1.09 10
3.93 10
7.06 10
2.64 102
0
1
0
Mn-54
Ru-103
2.69 10
1.64 10
1.97 10
1.08 101
0
1
0
Mn-56
Ru-105
5.05 10
2.45 10
3.96 10
1.82 101
0
1
2
Fe-59
Rh-103m
3.64 10
8.82 10
2.81 10
5.00 104
0
1
1
Co-56
Rh-105
9.86 10
2.62 10
8.39 10
1.73 102
Co-57
Rh-106
3.88 101
6.90 101
2.53 102
4.75 102
0
1
0
Co-58
Ag-110m
3.17 10
8.76 10
2.28 10
6.14 101
0
1
2
Co-60
Ag-111
7.59 10
8.91 10
5.90 10
5.90 103
0
1
1
Ni-65
Sn-117m
1.65 10
5.52 10
1.31 10
3.41 102
0
1
1
Zn-65
Sn-126
1.82 10
1.92 10
1.39 10
1.18 102
0
2
0
Zn-69m
Sb-124
1.41 10
5.67 10
9.58 10
4.25 101
0
2
0
Se-75
Sb-125
1.30 10
1.48 10
8.68 10
9.97 102
0
1
0
Br-84
Sb-126
4.90 10
9.07 10
4.18 10
6.66 101
1
2
0
Rb-86
Sb-127
2.94 10
2.20 10
2.25 10
1.61 101
0
1
1
Sr-92
Sb-128
4.01 10
1.01 10
3.14 10
7.20 101
0
1
0
Y-90m
Sb-129
2.10 10
4.58 10
1.42 10
3.36 101
2
4
1
Y-91
Sb-130
1.10 10
1.04 10
8.50 10
7.63 101
0
1
1
Y-91m
Te-123m
1.77 10
4.96 10
1.23 10
3.16 102
1
2
1
Y-92
Te-125m
7.91 10
1.87 10
5.94 10
9.07 103
1
2
2
Y-93
Te-127
2.69 10
1.65 10
2.06 10
1.11 103
Zr-95
Te-127m
2.40 100
5.91 102
1.73 101
2.78 103
1
2
1
Te-129
Zr-97
2.07 10
6.06 10
4.14 10
1.38 102
Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]
Radionuclide

Te-129m
Te-131m
Te-132
Te-133m
Te-134
I-129
I-130
I-131
I-132
I-133
I-134
I-135
Cs-134
Cs-134m
Cs-136
Cs-138
Ba-137m
Ba-139
Ba-140
La-140
La-141
La-142
Ce-141
Ce-143
Ce-144
Pr-145
Nd-147
Pm-148
Pm-148m
Pm-149
Pm-151
Eu-152
Eu-152m
Eu-154
Eu-155
Eu-156
Hf-181

Landolt-Brnstein
New Series VIII/4

6 External dosimetry
Source in soil
at a depth of
0.5 g cm2
Kerma rate
[(nGy h1)/
(kBq m2)]
1.65 101
4.51 100
8.05 101
5.79 100
2.84 100
1.14 101
7.05 100
1.29 100
7.35 100
2.01 100
8.26 100
4.79 100
5.09 100
1.08 101
6.75 100
6.96 100
1.98 100
1.45 101
6.21 101
6.93 100
1.27 101
6.57 100
2.44 101
9.28 101
6.37 102
8.32 102
4.71 101
1.78 100
6.55 100
1.23 103
1.09 100
3.53 100
9.47 101
3.85 100
1.88 101
3.98 100
1.79 100

Volume
source in air
Kerma rate
[(nGy h1)/
(kBq m3)]
9.04 103
3.22 101
5.15 102
5.36 101
2.03 101
6.01 103
4.93 101
8.68 102
5.26 101
1.40 101
6.05 101
3.67 101
3.64 101
5.90 103
5.08 101
5.44 101
1.40 101
9.18 103
4.14 102
5.44 101
9.97 103
6.26 101
1.59 102
6.34 102
4.36 103
3.11 103
3.11 102
1.35 101
4.64 101
2.36 103
6.88 102
1.82 101
6.91 102
2.89 101
1.22 102
3.09 101
1.25 101

Radionuclide

Ta-182
W-187
Pb-210
Pb-212
Bi-212
Ra-224
Ra-226
Ac-228
Th-228
Th-231
Th-232
Th-234
Pa-233
U-232
U-234
U-235
U-236
U-237
U-238
Np-237
Np-238
Np-239
Pu-236
Pu-238
Pu-239
Pu-240
Pu-242
Am-241
Am-242
Am-242m
Am-243
Cm-242
Cm-243
Cm-244
Cm-245
Cm-247

6-19
Source in soil
at a depth of
0.5 g cm2
Kerma rate
[(nGy h1)/
(kBq m2)]
4.00 100
1.70 100
2.71 102
4.74 101
3.38 101
3.33 102
2.19 102
3.05 100
1.43 102
1.31 101
7.89 103
3.33 102
7.37 101
1.06 102
9.47 103
5.55 101
7.90 103
4.80 101
8.33 103
1.42 101
2.05 100
6.02 101
1.08 102
1.06 102
4.38 103
9.25 103
8.56 103
1.12 101
6.76 102
3.25 102
1.81 101
9.38 103
4.47 101
8.87 103
3.10 101
1.03 100

Volume
source in air
Kerma rate
[(nGy h1)/
(kBq m3)]
2.99 101
1.09 101
1.51 103
3.24 102
4.36 102
2.17 103
1.46 103
2.19 101
8.75 104
7.02 103
4.54 104
2.07 103
4.54 102
7.24 104
5.80 104
3.34 102
5.44 104
3.11 102
6.88 104
8.42 103
1.31 101
3.78 102
7.06 104
6.19 104
2.41 104
5.83 104
5.04 104
7.96 103
4.39 103
1.61 103
1.14 102
6.05 104
2.97 102
5.62 104
2.07 102
7.16 102

6-20

6 External dosimetry

[Ref. p. 6-42

6.4 Conversion coefficients for neutrons


Significant radiation exposures to neutrons occur primarily at workplaces and not in the environment. In
the natural environment neutrons are found mainly in secondary cosmic ray fields [00Pel, 02Roe];
accidental exposures to neutron emitting radionuclides in clouds or on the soil are extremely unlikely and
therefore not considered here.
Neutron fields are in practice mixed radiation fields of wide neutron energy range, almost always
associated with photons. To obtain the conversion coefficients for such fields, appropriate averaging of
coefficients over the relevant spectra should be performed. The calculation of deposition of energy at any
point in a body resulting from external exposure in mixed fields is a complex process of summation over
all primary and secondary particle deposition.
Several authors calculated for incident neutrons the protection quantities organ absorbed dose and
effective dose using anthropomorphic phantoms such as the hermaphrodite MIRD-5 phantom or the sexspecific MIRD-type phantoms Adam and Eva (see Sect. 6.2 and 6.3). For the operational quantities, the
ICRU sphere and slab phantoms were used. Several Monte Carlo codes were applied such as the MCNP
[91Bri], SAM-CE [79Lic], MORSE-CG [75Emm], the JAERI code [Yam93], the PTB-code [90Hol] etc.
Extensive tables of organ dose conversion coefficients data are given in ICRP Report 74 [96ICR] and
ICRU Report 57 [978ICR] derived as sets of best estimates of the data of various authors. Table 6.7
lists the effective dose per unit of neutron fluence for idealized whole-body irradiation geometries and for
energies ranging from thermal up to 180 MeV. In the same table, the coefficients for ambient dose
equivalent are also given.
Table 6.7. Effective dose per unit neutron fluence E/ for monoenergetic neutrons incident in
various geometries on an adult anthropomorphic computational model. The last column of the table
shows the coefficients for ambient dose equivalent; [978ICR].
Energy
E/ [pSv cm2]
H*(10)/
[MeV]
AP
PA
RLAT
LLAT
ROT
ISO
[pSv cm2]
5.24
3.52
1.36
1.68
2.99
2.99
6.60
1.0 109
6.55
4.39
1.70
2.04
3.72
2.89
9.00
1.0 108
7.60
5.16
1.99
2.31
4.40
3.30
10.6
2.5 108
9.95
6.77
2.58
2.86
5.75
4.13
12.9
1.0 107
11.2
7.63
2.92
3.21
6.43
4.59
13.5
2.0 107
12.8
8.76
3.35
3.72
7.27
5.20
13.6
5.0 107
13.8
9.55
3.67
4.12
7.84
5.63
13.3
1.0 106
14.5
10.2
3.89
4.39
8.31
5.96
12.9
2.0 106
15.0
10.7
4.08
4.66
8.72
6.28
12.0
5.0 106
15.1
11.0
4.16
4.80
8.90
6.44
11.3
1.0 105
15.1
11.1
4.20
4.89
8.92
6.51
10.6
2.0 105
5
14.8
11.1
4.19
4.95
8.82
6.51
9.90
5.0 10
4
14.6
11.0
4.15
4.95
8.69
6.45
9.40
1.0 10
4
14.4
10.9
4.10
4.92
8.56
6.32
8.90
2.0 10
4
14.2
10.7
4.03
4.86
8.40
6.14
8.30
5.0 10
14.2
10.7
4.00
4.84
8.34
6.04
7.90
1.0 103
14.4
10.8
4.00
4.87
8.39
6.05
7.70
2.0 103
15.7
11.6
4.29
5.25
9.06
6.52
8.00
5.0 103
18.3
13.5
5.02
6.14
10.6
7.70
10.5
1.0 102
23.8
17.3
6.48
7.95
13.8
10.2
16.6
2.0 102
29.0
21.0
7.93
9.74
16.9
12.7
23.7
3.0 102
38.5
27.6
10.6
13.1
22.7
17.3
41.1
5.0 102
47.2
33.5
13.1
16.1
27.8
21.5
60.0
7.0 102

Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]

6 External dosimetry

Energy
[MeV]
1.0 101
1.5 101
2.0 101
3.0 101
5.0 101
7.0 101
9.0 101
1.0 100
1.2 100
2.0 100
3.0 100
4.0 100
5.0 100
6.0 100
7.0 100
8.0 100
9.0 100
1.0 101
1.2 101
1.4 101
1.5 101
1.6 101
1.8 101
2.0 101
3.0 101
5.0 101
7.5 101
1.0 102
1.3 102
1.5 102
1.8 102
2.0 102
a
Not available

E/ [pSv cm2]
RLAT
LLAT
16.4
20.1
21.2
25.5
25.6
30.3
33.4
38.6
46.8
53.2
58.3
66.6
69.1
79.6
74.5
86.0
85.8
99.8
129
153
171
195
198
224
217
244
232
261
244
274
253
285
261
294
268
302
278
315
286
324
290
328
293
331
299
335
305
338
324
naa
358
naa
397
naa
433
naa
467
naa
501
naa
542
naa

AP
59.8
80.2
99.0
133
188
231
267
282
310
383
432
458
474
483
490
494
497
499
499
496
494
491
486
480
458
437
429
429
432
438
445

PA
41.3
52.2
61.5
77.1
103
124
144
154
175
247
308
345
366
380
391
399
406
412
422
429
431
433
435
436
437
444
459
477
495
514
535

6-21

ROT
34.8
45.4
54.8
71.6
99.4
123
144
154
173
234
283
315
335
348
358
366
373
378
385
390
391
393
394
395
395
404
422
443
465
489
517

ISO
27.2
35.2
42.4
54.7
75.0
92.8
108
116
130
178
220
250
272
282
290
297
303
309
322
333
338
342
345
343
naa
naa
naa
naa
naa
naa
naa

H*(10)/
[pSv cm2]
88.0
132
170
233
322
375
400
416
425
420
412
408
405
400
405
409
420
440
480
520
540
555
570
600
515
400
300
285
260
245
250
260

6.5 Conversion coefficients for electrons


6.5.1 Occupational exposure
Unshielded whole body irradiation by monoenergetic electrons does not represent a practical situation in
occupational exposures and evaluated absorbed doses for electron beams are still sparse. However,
irradiation of the skin, the lens of the eye and other superficial organs are of concern in radiological
protection for electron energies below 10 MeV because the electron range is small, varying from 50 m to
about 5 cm for electron energies from 60 keV to 10 MeV. Table 6.8 shows some conversion coefficients
for organ absorbed doses determined with the MCNP-4 code for the MIRD-type phantoms Adam and
Eva, for monoenergetic electrons in the energy range of 100 keV to 10 MeV, incident in the AP geometry
Landolt-Brnstein
New Series VIII/4

6-22

6 External dosimetry

[Ref. p. 6-42

(Schultz and Zoetelief, data from [96ICR]). ICRU Report 43 contains dose distributions in
anthropomorphic phantoms resulting from irradiation by electrons of energies between 5 and 46 MeV
[88ICR].
Various workers performed Monte Carlo calculations with different codes (EGS4 [85Nel], MCNP-4
[91Bri], MCNP-BO code [94Gua1, 94Gua2], PTB-BG code [86Gro] etc.) enabling them to derive
fluence-to-dose-equivalent conversion coefficients for parallel electron beams of energies between
60 keV and 10 MeV. The conversion coefficients for H(0.07,), H(3,), Hp,slab(0.07,), Hp,slab(3,) and
Hp,slab(10,) were determined with the ICRU sphere or the 4-element ICRU tissue slab phantom,
respectively. A compilation of data can be found in [96Cha], in ICRP 74 and ICRU 57 [96ICR, 98ICR].
By appropriately averaging of these data, reference fluence-to-dose-equivalent conversion coefficients
were derived as a function of energy for normally incident electrons. Table 6.9 shows these data for
depths of 0.07 mm and 3 mm.
Table 6.8. Organ absorbed dose per unit fluence DT/ and effective dose per unit fluence E/ for
monoenergetic electrons incident in the AP geometry on an adult anthropomorphic computational model
(Schultz and Zoetelief, data from [96ICR]).
Energy [MeV]

0.1

0.4

0.6

Skin
Testes
Bone marrow
Stomach
Breast
Liver
Thyroid
Effective dose

98

171
0
0
0

0.1

1.5

1.0
1.5
2.0
2
DT/ and E/ [pGy cm ]
164
158
153
1
14
37
1
5
11
0
14
43
75

2.7

5.9

0
11

4.0

10.0

150
214
28
3
200
0
121
44

165
345
52
184
325
97
297
131

Table 6.9. Reference conversion coefficients from fluence to directional dose equivalent for monoenergetic electrons and normal incidence
Energy
H(0.07,0)/
Energy
H(0.07,0)/
H(3,0)/
H(3,0)/
[MeV]
[MeV]
[nSv cm2]
[nSv cm2]
[nSv cm2]
[nSv cm2]
1.00
0.312
0.301
0.07
0.221
1.25
0.296
0.486
0.08
1.056
1.50
0.287
0.524
0.09
1.527
1.75
0.282
0.512
0.10
1.661
2.00
0.279
0.481
0.1125
1.627
2.50
0.278
0.417
0.125
1.513
3.00
0.276
0.373
0.15
1.229
3.50
0.274
0.351
0.20
0.834
4.00
0.272
0.334
0.30
0.542
5.00
0.271
0.317
0.40
0.455
6.00
0.271
0.309
0.50
0.403
7.00
0.271
0.306
0.60
0.366
8.00
0.271
0.305
0.70
0.344
0.000
10.0
0.275
0.303
0.80
0.329
0.045

Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]

6 External dosimetry

6-23

6.5.2 Environmental exposure


Due to the short ranges of electrons emitted by radionuclides, electrons contribute only to the dose to
skin. Skin dose coefficients for a series of monoenergetic electron sources were calculated by Eckerman
and Ryman [93Eck1] using the code DOSFACTER of Kocher [88DOE]. The results are shown in Figs.
6.13 and 6.14 for submersion in contaminated air and for exposure to contaminated soil respectively.
These data can be then convoluted to the spectra of the various radionuclides, using the energy and
intensity of beta and electron emissions of radionuclides to obtain radionuclide specific conversion
coefficients.
100

10-1
Contaminated air

Contaminated soil

10-2
-1
-3
h(skin) [pSv s per Bq m ]

-1
-3
h(skin) [pSv s per Bq m ]

10-1
10-2
10-3
10-4

10-3
10-4
10-5
10-6
10-7
Infinite volume
Surface

10-8

10-5
10-9
10-6
10-2

10-1

100

101

Electron Energy [MeV]

Fig. 6.13. Electron skin dose coefficient for submersion


in air; [93Eck1].

10-10
10-2

10-1

100

Electron Energy [MeV]

Fig. 6.14. Electron skin dose coefficient for exposure to


contaminated soil on the surface and in the volume;
[93Eck1].

6.6 Doses from external exposure of radionuclides in the environment


Only photons, including bremsstrahlung, and electrons emitted by the radionuclides are sufficiently
penetrating to traverse the overlying tissues of the body and contribute to the dose to tissues and organs of
the body. The energy spectra of emitted radiation are either discrete, as in the case of photons, or
continuous, as in the case of beta particles and bremsstrahlung.
The dose coefficient H TS for tissue T for any exposure mode S can be expressed as
H TS =

[ y j ( E i ) H TS, j ( Ei ) + 0

j =e ,

y j ( E ) H TS, j ( E )dE ]

where y j ( Ei ) is the yield of radiations of type j and discrete energy Ei and y j (E ) denotes the yield of
radiations per nuclear transformation with continuous energy between E and E + dE. The other
summation is over all electron and photon radiations. The contribution of the radiations to the dose in
tissue or organ T is defined by the quantity H TS which is estimated by means of Monte Carlo calculations
and is given as a function of energy for tissue and organ T for each exposure mode [93Eck1].
Landolt-Brnstein
New Series VIII/4

6-24

6 External dosimetry

[Ref. p. 6-42

By using the dose conversion coefficients for monoenergetic sources of photon and electron radiation
and by scaling them to the emissions of the radionuclides of interest, dose coefficients from radionuclides
in the environment can be derived. The following tables contain data from the American Federal
Guidance Report No. 12, based on Monte Carlo radiation transport calculations and data obtained from
Eckerman [02Eck]. Tables 6.10 and 6.11 give the skin dose and effective dose coefficients for several
radionuclides for exposure to contaminated ground surface to a depth of 5 cm and for air submersion
respectively. The nuclear decay data used are from Eckerman et. al. [93Eck2] and are based on the ICRP
Publication 38 [83ICR] on radionuclide transformations.
Table 6.10. Effective dose and skin dose coefficients for exposure
depth of 5 cm ; [93Eck1] and [02Eck].
Effective dose
Radionuclide Skin dose
Radionuclide
[Sv/(Bq s m2)] [Sv/(Bq s m2)]
He-3
0.00
0.00
Ti-45
Be-7
Sc-46
1.041018
8.551019
Be-10
Ca-47
4.211021
3.021020
C-11
2.231017
1.771017
Sc-47
N-13
2.341017
V-47
1.771017
C-14
1.211022
Cr-48
5.501023
O-15
2.721017
Sc-48
1.781017
F-18
V-48
2.161017
1.771017
Ne-19
Ca-49
3.141017
1.791017
17
17
Na-22
4.4810
3.7610
Cr-49
Na-24
8.021017
Sc-49
6.801017
Al-26
5.521017
V-49
4.541017
Cr-51
Al-28
5.001017
3.011017
Mg-28
Mn-51
2.741017
2.311017
P-30
Fe-52
4.101017
1.811017
Si-31
Mn-52m
3.591018
7.881020
P-32
Mn-52
5.181018
9.051020
Si-32
Mn-53
2.671022
1.431022
Mn-54
P-33
4.111022
2.361022
Co-55
S-35
1.301022
6.081023
Fe-55
Cl-36
1.761019
9.721021
Co-56
Ar-37
0.00
0.00
Mn-56
Cl-38
5.041017
2.531017
Ni-56
K-38
7.711017
5.381017
Co-57
Ar-39
2.291020
3.371021
Ni-57
Cl-39
3.631017
2.461017
Co-58m
K-40
6.111018
2.701018
Co-58
Ar-41
2.751017
2.191017
Fe-59
Ca-41
0.00
0.00
K-42
Ni-59
2.521017
5.051018
Co-60m
K-43
2.081017
1.681017
17
17
Co-60
Sc-43
2.3810
1.9010
17
17
Cu-60
K-44
6.3810
3.8110
18
18
Fe-60
Sc-44m
5.8410
4.7910
17
17
Co-61
Sc-44
4.7610
3.6810
18
18
Cu-61
Ti-44
1.9010
1.4710
22
22
Co-62m
Ca-45
4.3010
2.5010
17
17
Cu-62
K-45
4.6610
3.1410

to contaminated ground surface to a


Skin dose
[Sv/(Bq s m2)]
1.941017
4.091017
2.271017
2.131018
2.801017
8.801018
6.791017
5.921017
6.471017
2.551017
7.491018
0.00
6.641019
3.051017
1.561017
6.241017
7.011017
0.00
1.721017
4.331017
0.00
7.091017
4.241017
3.531017
2.211018
3.851017
4.581023
2.021017
2.391017
0.00
9.161020
5.021017
8.681017
1.081022
3.261018
1.861017
6.561017
3.771017

Effective dose
[Sv/(Bq s m2)]
1.511017
3.451017
1.811017
1.741018
1.741017
7.191018
5.731017
4.991017
5.081017
1.801017
1.471019
0.00
5.431019
1.741017
1.261017
4.151017
5.921017
0.00
1.441017
3.441017
0.00
6.031017
2.881017
2.941017
1.811018
3.261017
9.551024
1.681017
2.031017
0.00
6.821020
4.271017
6.621017
4.601023
1.171018
1.431017
4.601017
1.771017
Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]
Radionuclide
Zn-62
Ni-63
Zn-63
Cu-64
Ga-65
Ni-65
Zn-65
Cu-66
Ga-66
Ge-66
Ni-66
Cu-67
Ga-67
Ge-67
Ga-68
Ge-68
As-69
Ge-69
Zn-69m
Zn-69
As-70
Ga-70
Se-70
As-71
Ge-71
Zn-71m
As-72
Ga-72
Zn-72
As-73
Ga-73
Se-73m
Se-73
As-74
Br-74m
Br-74
Kr-74
Br-75
Ge-75
Se-75
As-76
Br-76
Kr-76
As-77
Br-77
Ge-77
Kr-77
Se-77m
Landolt-Brnstein
New Series VIII/4

6 External dosimetry
Skin dose
[Sv/(Bq s m2)]
9.031018
0.00
3.271017
4.001018
3.211017
1.621017
1.181017
1.401017
6.181017
1.421017
3.111022
2.241018
3.011018
4.621017
2.651017
4.591022
3.761017
1.871017
8.791018
4.901019
9.171017
4.651018
2.341017
1.191017
4.651022
3.561017
4.931017
5.691017
2.811018
6.271020
8.131018
6.341018
2.421017
1.681017
1.001016
1.001016
3.161017
2.931017
2.111018
7.901018
2.151017
6.091017
8.861018
2.641019
6.591018
2.751017
2.621017
1.671018

Effective dose
[Sv/(Bq s m2)]
7.411018
0.00
1.921017
3.281018
2.021017
9.421018
9.961018
1.671018
4.131017
1.151017
1.731022
1.821018
2.451018
2.441017
1.661017
6.941024
1.781017
1.501017
7.211018
1.281020
7.021017
2.081019
1.701017
9.701018
7.021024
2.691017
3.111017
4.581017
2.291018
4.031020
5.311018
4.221018
1.841017
1.311017
6.921017
7.531017
2.001017
2.101017
6.111019
6.461018
7.641018
4.461017
7.231018
1.541019
5.421018
1.871017
1.741017
1.361018

Radionuclide
As-78
Ge-78
Kr-79
Rb-79
Se-79
Br-80m
Br-80
Rb-80
Sr-80
Kr-81m
Kr-81
Rb-81m
Rb-81
Se-81m
Se-81
Sr-81
Br-82
Rb-82m
Rb-82
Sr-82
Br-83
Kr-83m
Rb-83
Se-83
Sr-83
Br-84
Rb-84
Kr-85m
Kr-85
Sr-85m
Sr-85
Rb-86
Y-86m
Y-86
Zr-86
Kr-87
Rb-87
Sr-87m
Y-87
Kr-88
Nb-88
Rb-88
Y-88
Zr-88
Nb-89b
Nb-89a
Rb-89
Sr-89

6-25
Skin dose
[Sv/(Bq s m2)]
4.381017
5.941018
5.271018
3.601017
1.611022
1.201019
2.551018
5.751017
1.481020
2.611018
1.241019
7.661020
1.331017
2.401019
4.151018
4.041017
5.431017
5.991017
4.231017
1.451020
6.771019
3.471021
1.051017
5.231017
1.691017
5.041017
1.911017
3.371018
1.671019
4.511018
1.061017
7.191018
4.531018
7.381017
5.591018
3.331017
8.931022
6.731018
9.471018
3.991017
1.001016
4.411017
5.271017
8.321018
4.331017
4.891017
5.261017
3.691018

Effective dose
[Sv/(Bq s m2)]
2.171017
4.771018
4.331018
2.351017
7.611023
4.751020
1.391018
2.231017
5.331022
2.121018
9.481020
5.281020
1.061017
1.911019
2.291019
2.401017
4.551017
5.001017
1.931017
5.241022
1.431019
1.351022
8.611018
4.151017
1.361017
2.991017
1.581017
2.561018
4.401020
3.681018
8.731018
1.711018
3.691018
6.121017
4.521018
1.361017
5.551022
5.511018
7.771018
3.251017
7.111017
1.131017
4.531017
6.801018
2.391017
3.351017
3.521017
6.671020

6-26
Radionuclide
Zr-89
Mo-90
Nb-90
Sr-90
Y-90m
Y-90
Sr-91
Y-91m
Y-91
Sr-92
Y-92
Mo-93m
Mo-93
Nb-93m
Tc-93m
Tc-93
Y-93
Zr-93
Nb-94
Ru-94
Tc-94m
Tc-94
Y-94
Nb-95m
Nb-95
Tc-95m
Tc-95
Y-95
Zr-95
Nb-96
Tc-96m
Tc-96
Nb-97m
Nb-97
Ru-97
Tc-97m
Tc-97
Zr-97
Nb-98
Tc-98
Mo-99
Rh-99m
Rh-99
Tc-99m
Tc-99
Pd-100
Rh-100
Mo-101

6 External dosimetry
Skin dose
[Sv/(Bq s m2)]
2.411017
1.711017
8.421017
1.411020
1.311017
9.851018
1.981017
1.121017
4.071018
2.691017
2.531017
4.521017
2.991020
5.261021
1.391017
2.871017
1.641017
0.00
3.261017
1.091017
4.631017
5.511017
4.701017
1.281018
1.591017
1.381017
1.631017
3.901017
1.541017
5.111017
9.381019
5.161017
1.521017
1.571017
4.731018
3.511020
3.441020
9.341018
5.851017
2.951017
4.451018
1.401017
1.221017
2.381018
7.941022
1.711018
5.491017
3.161017

Effective dose
[Sv/(Bq s m2)]
2.001017
1.381017
7.061017
2.721021
1.071017
1.741019
1.211017
9.191018
1.321019
2.281017
4.711018
3.831017
2.231021
3.941022
1.201017
2.461017
1.781018
0.00
2.721017
9.011018
3.191017
4.591017
1.951017
1.031018
1.321017
1.141017
1.361017
1.521017
1.281017
4.261017
7.711019
4.301017
1.261017
1.141017
3.831018
7.181021
3.021021
3.171018
4.181017
2.451017
2.581018
1.161017
9.991018
1.951018
4.941022
1.291018
4.641017
2.331017

Radionuclide
Pd-101
Rh-101m
Rh-101
Tc-101
Ag-102
Rh-102m
Rh-102
Ag-103
Pd-103
Rh-103m
Ru-103
Ag-104m
Ag-104
Cd-104
Tc-104
Ag-105
Rh-105
Ru-105
Ag-106m
Ag-106
Rh-106m
Rh-106
Ru-106
Cd-107
Rh-107
Pd-107
Ag-108m
Ag-108
Ag-109m
Cd-109
In-109
Pd-109
Ag-110m
Ag-110
In-110b
In-110a
Sn-110
Ag-111
In-111
Sn-111
Ag-112
In-112
Cd-113m
Cd-113
In-113m
Sn-113
In-114m
In-114

[Ref. p. 6-42
Skin dose
[Sv/(Bq s m2)]
6.601018
6.221018
5.091018
9.091018
7.721017
1.081017
4.421017
1.721017
5.501020
6.221021
9.871018
2.981017
5.521017
4.801018
6.221017
1.071017
1.651018
1.761017
5.771017
1.931017
6.011017
2.361017
0.00
2.781019
8.301018
0.00
3.381017
4.491018
8.321020
1.461019
1.351017
9.381019
5.641017
1.521017
6.271017
3.851017
6.031018
1.361018
7.981018
1.171017
3.221017
6.561018
1.721020
7.141022
5.341018
1.811019
1.811018
5.581020

Effective dose
[Sv/(Bq s m2)]
5.391018
5.051018
4.121018
5.781018
5.731017
8.281018
3.671017
1.291017
8.911021
9.091022
8.121018
2.011017
4.591017
3.901018
3.401017
8.721018
1.341018
1.361017
4.831017
1.231017
5.011017
3.911018
0.00
1.621019
5.401018
0.00
2.791017
3.771019
4.611020
5.671020
1.121017
7.581020
4.731017
7.961019
5.231017
2.671017
4.871018
4.711019
6.451018
8.581018
1.161017
4.561018
2.541021
4.451022
4.361018
9.771020
1.461018
4.681020

Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]
Radionuclide
Ag-115
Cd-115m
Cd-115
In-115m
In-115
Sb-115
In-116m
Sb-116m
Sb-116
Te-116
Cd-117m
Cd-117
In-117m
In-117
Sb-117
Sn-117m
Sb-118m
In-119m
In-119
Sb-119
Sn-119m
I-120m
I-120
Sb-120b
Sb-120a
Xe-120
I-121
Sn-121m
Sn-121
Te-121m
Te-121
Xe-121
I-122
Sb-122
Xe-122
I-123
Sn-123m
Sn-123
Te-123m
Te-123
Xe-123
I-124
Sb-124n
Sb-124m
Sb-124
Cs-125
I-125
Sb-125
Landolt-Brnstein
New Series VIII/4

6 External dosimetry
Skin dose
[Sv/(Bq s m2)]
2.621017
4.441018
5.531018
3.281018
3.091021
2.041017
4.991017
6.381017
4.721017
9.181019
4.071017
2.481017
4.301018
1.451017
3.331018
2.831018
5.171017
1.211017
2.001017
1.081019
5.181020
1.241016
7.521017
4.941017
1.161017
8.291018
8.621018
2.611020
1.161021
4.221018
1.181017
4.221017
3.361017
1.291017
1.071018
3.041018
4.541018
2.991018
2.701018
9.891020
1.351017
2.381017
3.501022
7.651018
3.891017
1.701017
2.241019
8.821018

Effective dose
[Sv/(Bq s m2)]
1.211017
4.481019
4.031018
2.651018
1.521021
1.561017
4.211017
5.331017
3.671017
6.641019
3.451017
1.861017
1.501018
1.181017
2.671018
2.261018
4.351017
3.381019
1.331017
2.411020
1.131020
9.051017
4.651017
4.151017
7.691018
6.721018
6.841018
7.641021
7.581022
3.411018
9.681018
3.041017
1.661017
7.701018
8.051019
2.411018
2.231018
1.691019
2.161018
2.491020
1.041017
1.861017
7.381023
6.101018
3.101017
1.151017
6.361020
7.211018

Radionuclide
Sn-125
Te-125m
Xe-125
Ba-126
Cs-126
I-126
Sb-126m
Sb-126
Sn-126
Cs-127
Sb-127
Sn-127
Te-127m
Te-127
Xe-127
Ba-128
Cs-128
I-128
Sb-128b
Sb-128a
Sn-128
Cs-129
I-129
Sb-129
Te-129m
Te-129
Xe-129m
Cs-130
I-130
Sb-130
Ba-131m
Ba-131
Cs-131
I-131
La-131
Sb-131
Te-131m
Te-131
Xe-131m
Cs-132
I-132m
I-132
La-132
Te-132
Ba-133m
Ba-133
I-133
Te-133m

6-27
Skin dose
[Sv/(Bq s m2)]
1.471017
1.941019
5.091018
3.061018
4.431017
9.691018
3.681017
6.021017
7.671019
8.441018
1.501017
4.281017
6.641020
1.811019
5.341018
1.231018
2.921017
8.511018
6.641017
5.081017
1.301017
5.421018
1.481019
3.151017
1.961018
3.951018
4.321019
1.441017
4.521017
7.321017
1.161018
9.051018
1.311019
8.031018
1.491017
4.151017
2.951017
1.421017
1.581019
1.441017
6.881018
4.991017
4.691017
4.431018
1.131018
7.711018
1.421017
5.261017

Effective dose
[Sv/(Bq s m2)]
5.491018
5.811020
4.071018
2.441018
1.921017
7.701018
2.691017
4.901017
5.821019
6.851018
1.191017
3.261017
2.061020
8.801020
4.281018
9.361019
1.571017
1.571018
5.351017
3.451017
1.061017
4.351018
5.111020
2.471017
5.441019
1.001018
2.331019
8.811018
3.701017
5.611017
9.031019
7.351018
4.201020
6.561018
1.111017
3.181017
2.431017
7.141018
8.131020
1.191017
5.401018
3.941017
3.411017
3.541018
8.701019
6.181018
1.051017
3.961017

6-28
Radionuclide
Te-133
Xe-133m
Xe-133
Ce-134
Cs-134m
Cs-134
I-134
La-134
Te-134
Ba-135m
Ce-135
Cs-135m
Cs-135
I-135
La-135
Xe-135m
Xe-135
Cs-136
Nd-136
Pr-136
Ba-137m
Ce-137m
Ce-137
Cs-137
La-137
Pr-137
Cs-138
La-138
Nd-138
Pr-138m
Pr-138
Xe-138
Ba-139
Ce-139
Nd-139m
Nd-139
Pr-139
Ba-140
La-140
Ba-141
Ce-141
La-141
Nd-141m
Nd-141
Pm-141
Sm-141m
Sm-141
Ba-142

6 External dosimetry
Skin dose
[Sv/(Bq s m2)]
2.671017
5.841019
5.591019
1.721019
3.571019
3.241017
5.801017
2.341017
1.841017
9.931019
3.741017
3.301017
2.871022
3.271017
3.761019
9.071018
5.601018
4.421017
5.381018
5.221017
1.271017
8.221019
3.511019
9.231020
1.521019
1.161017
6.151017
2.451017
5.011019
5.201017
3.321017
2.811017
9.721018
2.781018
3.201017
9.731018
2.321018
4.231018
4.891017
2.621017
1.391018
1.091017
1.611017
1.181018
2.291017
4.461017
3.771017
2.291017

Effective dose
[Sv/(Bq s m2)]
1.611017
4.261019
4.001019
6.731020
2.641019
2.691017
4.521017
1.211017
1.501017
7.601019
3.071017
2.741017
1.551022
2.681017
2.371019
7.371018
4.231018
3.701017
4.251018
3.601017
1.031017
6.181019
2.141019
3.621021
5.591020
8.371018
4.021017
2.091017
3.311019
4.231017
1.431017
1.901017
8.291019
2.191018
2.641017
6.721018
1.741018
3.071018
3.931017
1.451017
1.121018
9.091019
1.311017
8.981019
1.281017
3.371017
2.411017
1.781017

Radionuclide
La-142
Pm-142
Pr-142m
Pr-142
Sm-142
Ce-143
La-143
Pm-143
Pr-143
Ce-144
Pm-144
Pr-144m
Pr-144
Eu-145
Gd-145
Pm-145
Pr-145
Sm-145
Eu-146
Gd-146
Pm-146
Sm-146
Eu-147
Gd-147
Nd-147
Pm-147
Pr-147
Sm-147
Tb-147
Eu-148
Gd-148
Pm-148m
Pm-148
Eu-149
Gd-149
Nd-149
Pm-149
Tb-149
Eu-150b
Eu-150a
Pm-150
Tb-150
Gd-151
Nd-151
Pm-151
Sm-151
Tb-151
Eu-152m
Eu-152

[Ref. p. 6-42
Skin dose
[Sv/(Bq s m2)]
6.111017
3.831017
0.00
8.941018
1.671018
6.971018
1.941017
6.171018
5.061019
3.331019
3.231017
1.031019
1.581017
2.911017
5.081017
2.441019
5.411018
5.421019
5.121017
3.821018
1.561017
0.00
9.631018
2.711017
2.761018
3.301022
2.391017
0.00
3.941017
4.471017
0.00
4.171017
1.811017
9.061019
8.141018
9.741018
1.151018
3.381017
3.091017
1.611018
3.581017
4.101017
8.521019
2.291017
6.951018
2.461023
1.791017
9.771018
2.311017

Effective dose
[Sv/(Bq s m2)]
4.591017
1.531017
0.00
1.121018
1.231018
4.501018
1.921018
5.041018
1.281020
2.611019
2.671017
5.181020
8.171019
2.441017
3.801017
1.211019
3.121019
2.851019
4.271017
2.931018
1.281017
0.00
7.901018
2.251017
2.081018
1.961022
1.441017
0.00
2.711017
3.711017
0.00
3.451017
9.941018
6.591019
6.591018
6.341018
2.021019
2.721017
2.541017
7.761019
2.451017
2.871017
6.081019
1.551017
5.301018
3.621024
1.461017
4.951018
1.931017
Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]
Radionuclide
Gd-152
Gd-153
Sm-153
Tb-153
Eu-154
Tb-154
Dy-155
Eu-155
Ho-155
Sm-155
Tb-155
Eu-156
Sm-156
Tb-156m
Tb-156n
Tb-156
Dy-157
Eu-157
Ho-157
Tb-157
Eu-158
Tb-158
Dy-159
Gd-159
Ho-159
Tb-160
Er-161
Ho-161
Tb-161
Ho-162m
Ho-162
Tm-162
Yb-162
Ho-164m
Ho-164
Dy-165
Er-165
Dy-166
Ho-166m
Ho-166
Tm-166
Yb-166
Ho-167
Tm-167
Yb-167
Er-169
Lu-169
Yb-169
Landolt-Brnstein
New Series VIII/4

6 External dosimetry
Skin dose
[Sv/(Bq s m2)]
0.00
1.331018
9.291019
4.031018
2.581017
4.511017
1.141017
9.061019
9.531018
4.931018
2.161018
2.931017
2.281018
2.511019
3.961020
3.641017
7.051018
6.101018
9.491018
2.231020
3.111017
1.591017
4.091019
1.381018
6.511018
2.291017
1.831017
5.951019
3.531019
1.111017
2.981018
3.861017
2.181018
4.321019
5.051019
2.261018
3.621019
4.941019
3.591017
5.661018
3.691017
9.431019
7.691018
2.441018
4.111018
8.531022
2.031017
5.021018

Effective dose
[Sv/(Bq s m2)]
0.00
9.541019
6.081019
3.201018
2.101017
3.881017
9.391018
7.061019
6.251018
1.481018
1.651018
2.251017
1.801018
1.551019
2.631020
3.051017
5.681018
4.021018
7.581018
1.221020
1.811017
1.321017
2.351019
7.531019
5.191018
1.911017
1.521017
3.671019
2.251019
9.191018
2.341018
2.961017
1.691018
2.611019
1.921019
4.161019
2.201019
3.491019
2.981017
4.971019
3.111017
6.351019
6.141018
1.891018
3.181018
5.431022
1.701017
3.891018

Radionuclide
Hf-170
Lu-170
Tm-170
Er-171
Lu-171
Tm-171
Er-172
Hf-172
Lu-172
Ta-172
Tm-172
Hf-173
Lu-173
Ta-173
Tm-173
Lu-174m
Lu-174
Ta-174
Hf-175
Ta-175
Tm-175
Yb-175
Lu-176m
Lu-176
Ta-176
W-176
Hf-177m
Lu-177m
Lu-177
Re-177
Ta-177
W-177
Yb-177
Hf-178m
Lu-178m
Lu-178
Re-178
Ta-178b
Ta-178a
W-178
Yb-178
Hf-179m
Lu-179
Ta-179
W-179
Hf-180m
Os-180
Re-180

6-29
Skin dose
[Sv/(Bq s m2)]
1.061017
4.791017
6.241019
8.551018
1.361017
7.021021
1.071017
1.401018
3.781017
3.561017
1.291017
7.591018
1.941018
1.451017
8.591018
7.571019
2.061018
1.541017
7.251018
1.801017
2.391017
8.071019
1.901018
9.981018
4.201017
2.471018
4.561017
1.991017
6.681019
1.471017
8.911019
1.761017
5.601018
4.851017
2.421017
9.381018
3.011017
2.031017
1.701018
1.521019
7.641019
1.811017
2.641018
3.561019
6.131019
2.051017
8.231019
2.441017

Effective dose
[Sv/(Bq s m2)]
8.571018
4.101017
6.501020
6.211018
1.121017
4.751021
8.791018
1.001018
3.161017
2.601017
8.071018
6.131018
1.461018
9.381018
6.671018
5.421019
1.651018
1.021017
5.851018
1.511017
1.811017
6.571019
1.781019
8.111018
3.561017
1.891018
3.711017
1.611017
5.391019
9.901018
6.601019
1.441017
3.141018
3.971017
1.841017
2.421018
2.011017
1.641017
1.351018
1.061019
6.001019
1.471017
5.481019
2.451019
4.061019
1.671017
6.041019
1.981017

6-30
Radionuclide
Ta-180m
Ta-180
Hf-181
Os-181
Re-181
W-181
Hf-182m
Hf-182
Ir-182
Os-182
Re-182b
Re-182a
Ta-182m
Ta-182
Hf-183
Ta-183
Hf-184
Ir-184
Re-184m
Re-184
Ta-184
Ir-185
Os-185
Ta-185
W-185
Ir-186a
Ir-186b
Pt-186
Re-186m
Re-186
Ta-186
Ir-187
Re-187
W-187
Ir-188
Pt-188
Re-188m
Re-188
W-188
Ir-189
Os-189m
Pt-189
Re-189
Ir-190n
Ir-190m
Ir-190
Os-190m
Ir-191m

6 External dosimetry
Skin dose
[Sv/(Bq s m2)]
5.751019
1.111017
1.131017
2.431017
1.531017
4.611019
1.891017
4.911018
3.971017
8.531018
3.701017
2.301017
4.491018
2.571017
1.701017
5.441018
5.341018
4.031017
7.481018
1.781017
3.441017
1.121017
1.461017
8.031018
2.341021
3.281017
2.081017
1.501017
1.691019
1.121018
4.131017
6.961018
0.00
1.031017
3.071017
3.571018
1.041018
7.881018
3.881020
1.191018
5.341022
6.001018
1.891018
3.201017
5.861022
2.951017
3.321017
1.111018

Effective dose
[Sv/(Bq s m2)]
4.001019
8.971018
9.291018
2.021017
1.261017
3.241019
1.551017
4.001018
2.291017
6.931018
3.091017
1.931017
3.591018
2.161017
1.271017
4.351018
3.901018
3.201017
6.131018
1.481017
2.751017
9.441018
1.201017
2.891018
1.661021
2.741017
1.611017
1.241017
1.151019
2.921019
2.671017
5.691018
0.00
8.051018
2.621017
2.851018
7.831019
1.051018
3.131020
9.091019
9.451024
4.871018
1.101018
2.631017
1.101023
2.421017
2.731017
8.751019

Radionuclide
Os-191m
Os-191
Pt-191
Ir-192m
Ir-192
Au-193
Hg-193m
Hg-193
Os-193
Pt-193m
Pt-193
Au-194
Hg-194
Ir-194m
Ir-194
Os-194
Tl-194m
Tl-194
Au-195m
Au-195
Hg-195m
Hg-195
Ir-195m
Ir-195
Pb-195m
Pt-195m
Tl-195
Hg-197m
Hg-197
Pt-197m
Pt-197
Tl-197
Au-198m
Au-198
Pb-198
Tl-198m
Tl-198
Au-199
Hg-199m
Pb-199
Pt-199
Tl-199
Au-200m
Au-200
Bi-200
Pb-200
Pt-200
Tl-200

[Ref. p. 6-42
Skin dose
[Sv/(Bq s m2)]
9.261020
1.181018
5.461018
3.161018
1.721017
2.671018
2.091017
3.471018
2.301018
1.421019
1.661021
2.111017
2.321021
4.901017
9.471018
9.621021
5.061017
1.591017
4.031018
1.111018
4.051018
3.691018
8.421018
1.491018
3.371017
9.941019
2.521017
1.581018
9.191019
1.411018
4.181019
7.931018
1.101017
9.071018
8.601018
2.461017
3.961017
1.671018
3.401018
2.941017
7.091018
4.651018
4.341017
1.231017
5.031017
3.641018
1.021018
2.641017

Effective dose
[Sv/(Bq s m2)]
6.741020
9.211019
4.381018
2.581018
1.411017
2.111018
1.731017
2.721018
1.171018
1.061019
3.381023
1.771017
5.711023
4.031017
1.681018
4.921021
3.961017
1.301017
3.271018
8.451019
3.291018
3.001018
6.671018
6.481019
2.701017
7.591019
2.111017
1.271018
7.001019
1.111018
3.011019
6.491018
8.911018
7.011018
7.001018
2.021017
3.361017
1.351018
2.741018
2.471017
3.501018
3.761018
3.581017
4.771018
4.071017
2.931018
7.901019
2.211017

Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]
Radionuclide
Au-201
Bi-201
Pb-201
Tl-201
Bi-202
Pb-202m
Pb-202
Tl-202
Bi-203
Hg-203
Pb-203
Po-203
Tl-204
Bi-205
Pb-205
Po-205
Bi-206
Tl-206
At-207
Bi-207
Po-207
Tl-207
Tl-208
Pb-209
Tl-209
Bi-210m
Bi-210
Pb-210
Po-210
At-211
Bi-211
Pb-211
Po-211
Bi-212
Pb-212
Po-212
Bi-213
Po-213
Bi-214
Pb-214
Po-214
At-215
Po-215
At-216
Po-216
At-217
At-218
Po-218
Landolt-Brnstein
New Series VIII/4

6 External dosimetry
Skin dose
[Sv/(Bq s m2)]
2.621018
2.761017
1.531017
1.371018
5.561017
4.231017
2.071021
9.401018
4.741017
4.921018
6.041018
3.371017
1.921019
3.341017
2.241021
3.181017
6.651017
3.031018
2.651017
3.211017
2.701017
2.481018
6.741017
3.801020
4.511017
5.321018
1.201018
2.221020
1.771022
5.731019
9.711019
3.081018
1.631019
7.851018
2.881018
0.00
4.531018
0.00
3.531017
5.231018
1.731021
4.071021
3.711021
2.251020
3.511022
6.401021
4.281020
1.891022

Effective dose
[Sv/(Bq s m2)]
9.301019
2.251017
1.261017
1.071018
4.611017
3.521017
3.931023
7.671018
4.011017
4.011018
4.891018
2.771017
1.771020
2.831017
4.391023
2.661017
5.571017
5.591020
2.221017
2.611017
2.251017
8.281020
5.581017
3.191021
3.431017
4.341018
2.431020
1.051020
1.471022
4.511019
7.931019
9.071019
1.351019
3.231018
2.341018
0.00
2.321018
0.00
2.571017
4.181018
1.441021
3.331021
3.051021
1.781020
2.921022
5.261021
2.501020
1.571022

Radionuclide
Rn-218
Fr-219
Rn-219
Fr-220
Rn-220
Fr-221
Fr-222
Ra-222
Rn-222
Ac-223
Fr-223
Ra-223
Ac-224
Ra-224
Ac-225
Ra-225
Ac-226
Ra-226
Th-226
Ac-227
Pa-227
Ra-227
Th-227
Ac-228
Pa-228
Ra-228
Th-228
Th-229
Pa-230
Th-230
U-230
Pa-231
Th-231
U-231
Np-232
Pa-232
Th-232
U-232
Np-233
Pa-233
U-233
Np-234
Pa-234m
Pa-234
Pu-234
Th-234
U-234
Np-235

6-31
Skin dose
[Sv/(Bq s m2)]
1.591020
7.251020
1.171018
2.001019
8.121021
6.221019
6.161018
1.931019
8.401021
8.911020
1.691018
2.501018
3.641018
2.031019
2.861019
9.801020
3.001018
1.321019
1.461019
2.451021
3.161019
4.341018
2.091018
2.151017
2.281017
0.00
3.811020
1.491018
1.301017
8.321021
2.381020
7.691019
2.151019
1.131018
2.441017
1.921017
5.041021
8.661021
1.491018
4.021018
8.261021
2.851017
8.021018
3.931017
1.091018
1.241019
5.611021
3.021020

Effective dose
[Sv/(Bq s m2)]
1.311020
5.941020
9.561019
1.611019
6.681021
5.061019
1.081019
1.581019
6.911021
7.011020
6.901019
2.031018
2.951018
1.661019
2.281019
4.581020
2.071018
1.071019
1.171019
1.801021
2.471019
2.621018
1.691018
1.651017
1.901017
0.00
2.851020
1.181018
1.091017
4.621021
1.561020
6.021019
1.411019
8.791019
2.021017
1.601017
2.051021
3.381021
1.201018
3.271018
4.811021
2.411017
3.331019
3.241017
8.731019
9.491020
1.551021
1.231020

6-32

6 External dosimetry
Skin dose
[Sv/(Bq s m2)]
3.271021
8.761019
4.221018
1.551018
8.921018
2.281017
1.501017
1.921017
4.401021
4.381021
2.461018
1.881018
6.611018
4.111021
3.341021
4.801023
6.951018
7.311019
1.861017
3.911021
0.00
7.781018
2.291018
1.641018
4.031021
3.931021
5.661022
9.071021
1.411018
1.271023
1.011017
1.011019
4.631021
6.141020
1.891018
2.081018
2.831020

Effective dose
[Sv/(Bq s m2)]
2.921022
4.941019
3.421018
1.251018
7.021018
1.741017
1.151017
1.591017
7.061022
4.431022
1.961018
1.521018
5.411018
4.691022
3.351022
1.861023
5.681018
3.391019
1.521017
4.451022
0.00
6.461018
1.841018
1.321018
6.601022
5.051022
3.041022
5.561021
1.141018
1.451024
8.041018
4.541020
9.121022
2.361020
1.521018
1.691018
9.021021

Table 6.11. Skin dose and effective dose coefficients for air submersion; [93Eck1].
Effective Dose
Radionuclide Skin Dose
Radionuclide Skin Dose
[Sv/(Bq s m2)] [Sv/(Bq s m2)]
[Sv/(Bq s m2)]
He-3
0.00
0.00
C-14
2.431016
15
15
Be-7
2.7410
2.1910
O-15
1.041013
14
16
Be-10
1.2910
1.3810
F-18
6.941014
14
14
C-11
7.9110
Ne-19
4.5610
1.211013
14
14
N-13
8.6810
Na-22
4.5710
1.331013

Effective Dose
[Sv/(Bq s m2)]
2.601018
4.591014
4.561014
4.621014
1.021013

Radionuclide
Pu-235
U-235
Np-236a
Np-236b
Pu-236
U-236
Am-237
Np-237
Pu-237
U-237
Am-238
Cm-238
Np-238
Pu-238
U-238
Am-239
Np-239
Pu-239
U-239
Am-240
Cm-240
Np-240m
Np-240
Pu-240
U-240
Am-241
Cm-241
Pu-241
Am-242m
Am-242
Cm-242
Pu-242
Am-243
Cm-243
Pu-243
Am-244m
Am-244
Cf-244
Cm-244

Skin dose
[Sv/(Bq s m2)]
1.521018
3.011018
2.121018
8.381019
6.041021
4.461021
7.211018
4.031019
7.741019
2.371018
1.791017
1.261018
1.191017
5.031021
3.541021
4.201018
3.171018
2.871021
2.101018
2.071017
5.871021
1.161017
2.691017
4.831021
2.641020
2.891019
9.941018
2.951023
2.051020
2.691019
5.421021
4.031021
7.591019
2.441018
3.911019
2.611018
1.651017
6.021021
4.891021

Effective dose
[Sv/(Bq s m2)]
1.221018
2.451018
1.691018
6.721019
8.631022
8.361022
5.881018
2.961019
6.121019
1.881018
1.491017
1.011018
9.411018
5.781022
4.201022
3.391018
2.571018
1.011021
6.371019
1.721017
6.011022
5.761018
2.211017
5.661022
5.791021
1.851019
8.121018
2.201023
6.371021
1.881019
6.381022
4.941022
5.781019
1.981018
3.011019
4.801020
1.351017
6.801022
4.791022

[Ref. p. 6-42

Radionuclide
Pu-244
Am-245
Bk-245
Cm-245
Pu-245
Am-246m
Am-246
Bk-246
Cf-246
Cm-246
Pu-246
Bk-247
Cm-247
Cf-248
Cm-248
Bk-249
Cf-249
Cm-249
Bk-250
Cf-250
Cm-250
Es-250
Cf-251
Es-251
Cf-252
Fm-252
Cf-253
Es-253
Fm-253
Cf-254
Es-254m
Es-254
Fm-254
Fm-255
Fm-257
Md-257
Md-258

Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]
Radionuclide
Na-24
Al-26
Al-28
Mg-28
P-30
Si-31
P-32
Si-32
P-33
S-35
Cl-36
Ar-37
Cl-38
K-38
Ar-39
Cl-39
K-40
Ar-41
Ca-41
K-42
K-43
Sc-43
K-44
Sc-44m
Sc-44
Ti-44
Ca-45
K-45
Ti-45
Sc-46
Ca-47
Sc-47
V-47
Cr-48
Sc-48
V-48
Ca-49
Cr-49
Sc-49
V-49
Cr-51
Mn-51
Fe-52
Mn-52m
Mn-52
Mn-53
Mn-54
Co-55
Fe-55
Landolt-Brnstein
New Series VIII/4

6 External dosimetry
Skin Dose
[Sv/(Bq s m2)]
2.751013
1.811013
1.881013
8.331014
1.561013
3.781014
4.491014
8.271016
1.381015
2.921016
1.471014
0.00
1.941013
2.661013
1.071014
1.361013
4.201014
1.011013
0.00
1.151013
7.111014
7.911014
2.351013
1.721014
1.581013
6.791015
1.461015
1.741013
7.071014
1.171013
8.021014
1.281014
1.081013
2.401014
2.011013
1.721013
2.461013
9.651014
5.431014
0.00
1.751015
1.181013
5.171014
2.131013
1.991013
0.00
4.671014
1.391013
0.00

Effective Dose
[Sv/(Bq s m2)]
2.081013
1.281013
8.871014
6.381014
4.681014
4.831016
5.361016
8.681018
1.451017
3.111018
1.661016
0.00
7.581014
1.561013
1.151016
6.901014
7.921015
6.141014
0.00
1.481014
4.351014
4.881014
1.141013
1.241014
9.871014
4.701015
1.531017
9.201014
3.891014
9.361014
5.061014
4.671015
4.491014
1.871014
1.571013
1.361013
1.661013
4.681014
7.161016
0.00
1.381015
4.511014
3.271014
1.131013
1.621013
0.00
3.831014
9.161014
0.00

Radionuclide
Co-56
Mn-56
Ni-56
Co-57
Ni-57
Co-58m
Co-58
Fe-59
Ni-59
Co-60m
Co-60
Cu-60
Fe-60
Co-61
Cu-61
Co-62m
Cu-62
Zn-62
Ni-63
Zn-63
Cu-64
Ga-65
Ni-65
Zn-65
Cu-66
Ga-66
Ge-66
Ni-66
Cu-67
Ga-67
Ge-67
Ga-68
Ge-68
As-69
Ge-69
Zn-69m
Zn-69
As-70
Ga-70
Se-70
As-71
Ge-71
Zn-71m
As-72
Ga-72
Zn-72
As-73
Ga-73

6-33
Skin Dose
[Sv/(Bq s m2)]
2.131013
1.511013
9.611014
6.631015
1.171013
3.051019
5.581014
7.131014
0.00
3.461016
1.451013
2.821013
1.641016
3.241014
6.501014
2.251013
1.441013
2.521014
0.00
1.231013
1.641014
1.191013
7.181014
3.291014
7.691014
2.111013
4.261014
1.011015
1.181014
8.501015
1.681013
1.011013
6.621018
1.431013
5.961014
2.441014
1.811014
2.891013
4.171014
8.361014
3.781014
6.711018
1.211013
1.701013
1.861013
1.001014
2.781016
4.371014

Effective Dose
[Sv/(Bq s m2)]
1.731013
8.161014
7.821014
4.971015
9.121014
6.061020
4.441014
5.621014
0.00
2.001016
1.191013
1.871013
1.791018
3.741015
3.721014
1.301013
4.601014
1.921014
0.00
5.001014
8.501015
5.281014
2.671014
2.721014
4.891015
1.231013
3.001014
1.061017
4.901015
6.491015
6.451014
4.291014
1.011019
4.611014
3.991014
1.841014
1.991016
1.921013
8.401016
4.401014
2.531014
1.021019
6.991014
8.261014
1.311013
6.171015
1.551016
1.391014

6-34
Radionuclide
Se-73m
Se-73
As-74
Br-74m
Br-74
Kr-74
Br-75
Ge-75
Se-75
As-76
Br-76
Kr-76
As-77
Br-77
Ge-77
Kr-77
Se-77m
As-78
Ge-78
Kr-79
Rb-79
Se-79
Br-80m
Br-80
Rb-80
Sr-80
Kr-81m
Kr-81
Rb-81m
Rb-81
Se-81m
Se-81
Sr-81
Br-82
Rb-82m
Rb-82
Sr-82
Br-83
Kr-83m
Rb-83
Se-83
Sr-83
Br-84
Rb-84
Kr-85m
Kr-85
Sr-85m
Sr-85

6 External dosimetry
Skin Dose
[Sv/(Bq s m2)]
2.391014
8.311014
5.801014
3.311013
3.401013
1.161013
1.011013
2.711014
2.161014
9.611014
1.971013
2.371014
1.201014
1.771014
1.021013
9.741014
6.991015
1.651013
2.751014
1.501014
1.281013
3.711016
7.131016
2.021014
2.111013
1.441016
9.421015
4.041016
4.011016
4.461014
1.401015
3.941014
1.441013
1.541013
1.681013
1.581013
1.421016
1.851014
3.561017
2.771014
1.691013
5.201014
1.881013
5.711014
2.241014
1.321014
1.231014
2.831014

Effective Dose
[Sv/(Bq s m2)]
1.091014
4.781014
3.401014
1.961013
2.261013
5.201014
5.431014
1.781015
1.681014
2.061014
1.261013
1.861014
5.091016
1.401014
4.981014
4.511014
3.631015
6.031014
1.231014
1.121014
6.081014
3.941018
2.371016
3.731015
5.771014
5.001018
5.561015
2.441016
1.631016
2.731014
5.481016
8.691016
6.241014
1.211013
1.341013
5.011014
4.921018
5.341016
1.201018
2.211014
1.141013
3.601014
9.021014
4.181014
6.871015
2.401016
9.481015
2.241014

Radionuclide
Rb-86
Y-86m
Y-86
Zr-86
Kr-87
Rb-87
Sr-87m
Y-87
Kr-88
Nb-88
Rb-88
Y-88
Zr-88
Nb-89b
Nb-89a
Rb-89
Sr-89
Zr-89
Mo-90
Nb-90
Sr-90
Y-90m
Y-90
Sr-91
Y-91m
Y-91
Sr-92
Y-92
Mo-93m
Mo-93
Nb-93m
Tc-93m
Tc-93
Y-93
Zr-93
Nb-94
Ru-94
Tc-94m
Tc-94
Y-94
Nb-95m
Nb-95
Tc-95m
Tc-95
Y-95
Zr-95
Nb-96
Tc-96m

[Ref. p. 6-42
Skin Dose
[Sv/(Bq s m2)]
4.851014
1.281014
2.171013
1.561014
1.371013
3.151015
2.151014
2.511014
1.351013
3.121013
1.831013
1.541013
2.261014
1.561013
1.631013
1.871013
3.691014
7.071014
5.521014
2.661013
9.201015
3.751014
6.241014
8.141014
3.111014
3.851014
8.561014
1.141013
1.321013
2.431016
4.281017
4.621014
8.301014
8.501014
0.00
9.521014
2.951014
1.551013
1.511013
1.801013
1.121014
4.301014
3.761014
4.421014
1.591013
4.501014
1.521013
2.681015

Effective Dose
[Sv/(Bq s m2)]
4.941015
9.591015
1.691013
1.171014
3.971014
3.301017
1.411014
1.991014
9.711014
1.891013
3.331014
1.301013
1.731014
6.621014
8.651014
1.011013
4.371016
5.311014
3.641014
2.051013
9.831017
2.771014
7.921016
3.271014
2.371014
6.221016
6.411014
1.321014
1.061013
1.731017
3.051018
3.531014
6.961014
5.281015
0.00
7.201014
2.361014
8.641014
1.221013
5.391014
2.741015
3.491014
2.991014
3.581014
4.661014
3.361014
1.141013
2.091015

Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]
Radionuclide
Tc-96
Nb-97m
Nb-97
Ru-97
Tc-97m
Tc-97
Zr-97
Nb-98
Tc-98
Mo-99
Rh-99m
Rh-99
Tc-99m
Tc-99
Pd-100
Rh-100
Mo-101
Pd-101
Rh-101m
Rh-101
Tc-101
Ag-102
Rh-102m
Rh-102
Ag-103
Pd-103
Rh-103m
Ru-103
Ag-104m
Ag-104
Cd-104
Tc-104
Ag-105
Rh-105
Ru-105
Ag-106m
Ag-106
Rh-106m
Rh-106
Ru-106
Cd-107
Pd-107
Rh-107
Ag-108m
Ag-108
Ag-109m
Cd-109
In-109
Landolt-Brnstein
New Series VIII/4

6 External dosimetry
Skin Dose
[Sv/(Bq s m2)]
1.401013
4.161014
6.511014
1.321014
5.551016
2.711016
5.551014
1.961013
8.531014
3.141014
3.941014
3.421014
7.141015
2.741015
6.111015
1.631013
1.141013
1.941014
1.711014
1.491014
4.771014
2.451013
3.681014
1.191013
5.841014
3.901016
4.491017
2.771014
1.001013
1.561013
1.381014
2.251013
2.901014
1.071014
6.731014
1.581013
7.271014
1.811013
1.091013
0.00
1.501015
0.00
4.421014
9.051014
4.001014
5.591016
9.951016
3.911014

Effective Dose
[Sv/(Bq s m2)]
1.141013
3.311014
2.991014
9.911015
3.721017
2.261017
8.901015
1.141013
6.411014
6.991015
3.061014
2.631014
5.251015
2.871017
3.981015
1.331013
6.481014
1.421014
1.291014
1.091014
1.501014
1.571013
2.151014
9.681014
3.431014
5.321017
6.021018
2.081014
5.481014
1.231013
1.041014
9.611014
2.261014
3.471015
3.561014
1.291013
3.181014
1.351013
1.061014
0.00
5.111016
0.00
1.411014
7.241014
1.251015
1.591016
2.281016
2.981014

Radionuclide
Pd-109
Ag-110m
Ag-110
In-110b
In-110a
Sn-110
Ag-111
In-111
Sn-111
Ag-112
In-112
Cd-113m
Cd-113
In-113m
Sn-113
In-114m
In-114
Ag-115
Cd-115m
Cd-115
In-115m
In-115
Sb-115
In-116m
Sb-116m
Sb-116
Te-116
Cd-117m
Cd-117
In-117m
In-117
Sb-117
Sn-117m
Sb-118m
In-119m
In-119
Sb-119
Sn-119m
I-120m
I-120
Sb-120b
Sb-120a
Xe-120
I-121
Sn-121m
Sn-121
Te-121m
Te-121

6-35
Skin Dose
[Sv/(Bq s m2)]
2.151014
1.571013
8.221014
1.711013
1.291013
1.661014
2.191014
2.291014
4.221014
1.331013
2.881014
8.481015
2.411015
2.181014
8.201016
1.051014
2.951015
1.111013
3.991014
2.971014
1.811014
6.181015
6.521014
1.581013
1.821013
1.501013
3.371015
1.291013
8.791014
3.171014
5.161014
1.031014
1.251014
1.461013
7.111014
8.201014
7.091016
3.421016
3.861013
2.551013
1.391013
4.461014
2.401014
2.721014
1.071015
3.711015
1.231014
3.181014

Effective Dose
[Sv/(Bq s m2)]
4.201016
1.271013
2.461015
1.391013
7.151014
1.251014
1.381015
1.681014
2.301014
3.231014
1.191014
9.061017
2.531017
1.121014
3.151016
3.891015
1.591016
3.461014
1.481015
1.051014
6.861015
6.551017
4.021014
1.181013
1.451013
1.021013
1.981015
9.891014
5.141014
4.071015
3.061014
7.151015
6.111015
1.191013
1.261015
3.531014
1.501016
7.041017
2.491013
1.311013
1.141013
2.001014
1.791014
1.781014
5.241017
3.901017
8.991015
2.501014

6-36
Radionuclide
Xe-121
I-122
Sb-122
Xe-122
I-123
Sn-123m
Sn-123
Te-123m
Te-123
Xe-123
I-124
Sb-124n
Sb-124m
Sb-124
Cs-125
I-125
Sb-125
Sn-125
Te-125m
Xe-125
Ba-126
Cs-126
I-126
Sb-126m
Sb-126
Sn-126
Cs-127
Sb-127
Sn-127
Te-127m
Te-127
Xe-127
Ba-128
Cs-128
I-128
Sb-128b
Sb-128a
Sn-128
Cs-129
I-129
Sb-129
Te-129m
Te-129
Xe-129m
Cs-130
I-130
Sb-130
Ba-131m

6 External dosimetry
Skin Dose
[Sv/(Bq s m2)]
1.401013
1.251013
6.031014
3.361015
9.401015
3.581014
3.281014
8.481015
6.321016
4.521014
7.391014
2.331018
2.461014
1.261013
5.971014
1.391015
2.651014
7.131014
1.941015
1.501014
9.261015
1.621013
3.371014
1.241013
1.731013
6.651015
2.381014
5.581014
1.411013
8.491016
1.141014
1.571014
3.851015
1.071013
5.381014
1.991013
1.731013
4.501014
1.521014
1.101015
1.051013
1.491014
3.571014
8.291015
5.481014
1.361013
2.291013
3.941015

Effective Dose
[Sv/(Bq s m2)]
8.621014
4.311014
2.021014
2.191015
6.491015
6.141015
6.981016
5.811015
1.511016
2.821014
5.041014
4.671019
1.581014
8.621014
3.011014
3.731016
1.871014
1.541014
3.351016
1.081014
6.411015
4.961014
2.011014
7.011014
1.281013
1.841015
1.781014
3.121014
9.031014
1.121016
3.341016
1.121014
2.541015
4.061014
4.331015
1.411013
9.081014
2.771014
1.131014
2.811016
6.711014
1.561015
2.861015
9.141016
2.301014
9.671014
1.501013
2.641015

Radionuclide
Ba-131
Cs-131
I-131
La-131
Sb-131
Te-131m
Te-131
Xe-131m
Cs-132
I-132m
I-132
La-132
Te-132
Ba-133m
Ba-133
I-133
Te-133m
Te-133
Xe-133m
Xe-133
Ce-134
Cs-134m
Cs-134
I-134
La-134
Te-134
Ba-135m
Ce-135
Cs-135m
Cs-135
I-135
La-135
Xe-135m
Xe-135
Cs-136
Nd-136
Pr-136
Ba-137m
Ce-137m
Ce-137
Cs-137
La-137
Pr-137
Cs-138
La-138
Nd-138
Pr-138m
Pr-138

[Ref. p. 6-42
Skin Dose
[Sv/(Bq s m2)]
2.551014
7.841016
2.981014
4.871014
1.401013
8.851014
6.891014
4.821015
3.921014
2.221014
1.581013
1.491013
1.391014
1.361014
2.191014
5.831014
1.741013
1.061013
1.041014
4.971015
9.601016
2.881015
9.451014
1.871013
8.881014
6.351014
1.301014
1.101013
9.101014
9.061016
1.111013
1.491015
2.971014
3.121014
1.251013
1.711014
1.691013
3.731014
1.201014
1.451015
8.631015
8.681016
4.011014
2.171013
7.091014
1.921015
1.521013
1.251013

Effective Dose
[Sv/(Bq s m2)]
1.921014
2.381016
1.691014
2.911014
8.841014
6.551014
1.921014
3.491016
3.111014
1.421014
1.051013
9.411014
9.321015
2.441015
1.621014
2.761014
1.071013
4.341014
1.281015
1.331015
3.521016
7.951016
7.061014
1.221013
3.151014
3.941014
2.161015
7.931014
7.251014
9.501018
7.541014
7.751016
1.901014
1.101014
9.941014
1.151014
9.721014
2.691014
1.831015
7.301016
9.281017
3.001016
2.201014
1.151013
5.841014
1.071015
1.131013
3.721014

Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]
Radionuclide
Xe-138
Ba-139
Ce-139
Nd-139m
Nd-139
Pr-139
Ba-140
La-140
Ba-141
Ce-141
La-141
Nd-141m
Nd-141
Pm-141
Sm-141m
Sm-141
Ba-142
La-142
Pm-142
Pr-142m
Pr-142
Sm-142
Ce-143
La-143
Pm-143
Pr-143
Ce-144
Pm-144
Pr-144m
Pr-144
Eu-145
Gd-145
Pm-145
Pr-145
Sm-145
Eu-146
Gd-146
Pm-146
Sm-146
Eu-147
Gd-147
Nd-147
Pm-147
Pr-147
Sm-147
Tb-147
Eu-148
Gd-148
Pm-148m
Landolt-Brnstein
New Series VIII/4

6 External dosimetry
Skin Dose
[Sv/(Bq s m2)]
1.071013
6.161014
8.941015
9.171014
3.501014
8.751015
2.521014
1.661013
1.071013
1.021014
6.581014
4.671014
4.241015
8.421014
1.391013
1.271013
8.371014
2.161013
1.441013
0.00
5.671014
6.441015
3.961014
9.641014
1.721014
1.761014
2.931015
8.711014
5.081016
8.431014
8.331014
1.661013
1.221015
4.441014
2.641015
1.431013
1.331014
4.641014
0.00
2.771014
7.671014
1.951014
8.111016
9.751014
0.00
1.271013
1.221013
0.00
1.181013

Effective Dose
[Sv/(Bq s m2)]
5.481014
2.541015
5.971015
7.121014
1.771014
4.751015
8.071015
1.111013
3.921014
3.101015
2.881015
3.451014
2.591015
3.391014
9.071014
6.441014
4.841014
1.371013
4.011014
0.00
3.501015
3.431015
1.211014
5.781015
1.351014
1.941016
7.631016
6.951014
2.201016
2.651015
6.781014
1.091013
5.491016
1.121015
1.261015
1.151013
8.611015
3.341014
0.00
2.141014
5.981014
5.721015
8.671018
3.901014
0.00
7.291014
9.831014
0.00
9.011014

Radionuclide
Pm-148
Eu-149
Gd-149
Nd-149
Pm-149
Tb-149
Eu-150b
Eu-150a
Pm-150
Tb-150
Gd-151
Nd-151
Pm-151
Sm-151
Tb-151
Eu-152m
Eu-152
Gd-152
Gd-153
Sm-153
Tb-153
Eu-154
Tb-154
Dy-155
Eu-155
Ho-155
Sm-155
Tb-155
Eu-156
Sm-156
Tb-156m
Tb-156n
Tb-156
Dy-157
Eu-157
Ho-157
Tb-157
Eu-158
Tb-158
Dy-159
Gd-159
Ho-159
Tb-160
Er-161
Ho-161
Tb-161
Ho-162m
Ho-162

6-37
Skin Dose
[Sv/(Bq s m2)]
7.971014
3.091015
2.421014
4.991014
2.191014
1.021013
8.501014
2.051014
1.341013
1.311013
3.251015
9.121014
3.321014
1.901019
5.071014
4.851014
6.901014
0.00
5.001015
1.451014
1.231014
8.291014
1.381013
3.271014
3.391015
3.461014
4.011014
7.291015
9.981014
1.461014
1.111015
3.561016
1.041013
1.941014
3.571014
2.901014
1.061016
1.211013
4.701014
1.891015
1.911014
1.981014
7.341014
5.231014
2.591015
7.691015
3.221014
1.011014

Effective Dose
[Sv/(Bq s m2)]
2.761014
1.951015
1.751014
1.681014
7.081016
7.511014
6.641014
2.221015
6.771014
7.751014
1.881015
4.211014
1.401014
2.461020
3.871014
1.361014
5.281014
0.00
3.111015
2.041015
8.861015
5.751014
1.141013
2.561014
2.141015
1.651014
4.431015
4.841015
6.381014
4.931015
6.241016
9.731017
8.341014
1.481014
1.091014
2.041014
5.341017
5.001014
3.581014
9.931016
2.161015
1.431014
5.191014
4.111014
1.401015
8.931016
2.541014
6.701015

6-38
Radionuclide
Tm-162
Yb-162
Ho-164m
Ho-164
Dy-165
Er-165
Dy-166
Ho-166m
Ho-166
Tm-166
Yb-166
Ho-167
Tm-167
Yb-167
Er-169
Lu-169
Yb-169
Hf-170
Lu-170
Tm-170
Er-171
Lu-171
Tm-171
Er-172
Hf-172
Lu-172
Ta-172
Tm-172
Hf-173
Lu-173
Ta-173
Tm-173
Lu-174m
Lu-174
Ta-174
Hf-175
Ta-175
Tm-175
Yb-175
Lu-176m
Lu-176
Ta-176
W-176
Hf-177m
Lu-177m
Lu-177
Re-177
Ta-177

6 External dosimetry
Skin Dose
[Sv/(Bq s m2)]
1.241013
6.991015
1.931015
8.331015
2.821014
1.611015
5.791015
9.901014
4.461014
1.081013
3.881015
2.951014
1.171014
1.381014
2.831015
5.901014
1.731014
3.001014
1.461013
1.811014
4.221014
3.801014
3.171017
3.211014
5.461015
1.071013
1.161013
5.761014
2.231014
6.451015
5.081014
3.891014
2.891015
6.531015
5.361014
2.151014
5.321014
9.101014
6.931015
2.721014
3.741014
1.251013
8.741015
1.391013
5.891014
7.131015
5.171014
3.361015

Effective Dose
[Sv/(Bq s m2)]
8.501014
4.921015
1.061015
8.031016
1.351015
8.961016
1.211015
7.841014
1.721015
8.781014
2.351015
1.591014
5.391015
9.481015
2.971017
4.751014
1.131014
2.291014
1.211013
3.671016
1.641014
3.001014
1.771017
2.291014
3.401015
8.641014
7.101014
2.301014
1.661014
4.421015
2.551014
1.721014
1.841015
4.941015
2.751014
1.541014
4.241014
4.811014
1.751015
7.651016
2.111014
1.031013
5.981015
9.671014
4.241014
1.501015
2.761014
2.151015

Radionuclide
W-177
Yb-177
Hf-178m
Lu-178m
Lu-178
Re-178
Ta-178b
Ta-178a
W-178
Yb-178
Hf-179m
Lu-179
Ta-179
W-179
Hf-180m
Os-180
Re-180
Ta-180m
Ta-180
Hf-181
Os-181
Re-181
W-181
Hf-182m
Hf-182
Ir-182
Os-182
Re-182b
Re-182a
Ta-182m
Ta-182
Hf-183
Ta-183
Hf-184
Ir-184
Re-184m
Re-184
Ta-184
Ir-185
Os-185
Ta-185
W-185
Ir-186a
Ir-186b
Pt-186
Re-186m
Re-186
Ta-186

[Ref. p. 6-42
Skin Dose
[Sv/(Bq s m2)]
5.111014
3.601014
1.361013
9.061014
5.681014
1.041013
5.871014
5.651015
6.091016
1.071014
5.261014
2.991014
1.451015
2.581015
5.821014
3.191015
7.111014
3.671015
3.261014
3.621014
7.031014
4.761014
1.841015
5.821014
1.461014
1.351013
2.461014
1.081013
6.711014
1.931014
7.851014
6.831014
2.621014
3.121014
1.211013
2.191014
5.001014
1.161013
3.521014
4.011014
5.201014
4.521015
9.551014
6.411014
4.101014
7.241016
2.031014
1.491013

Effective Dose
[Sv/(Bq s m2)]
3.911014
8.821015
1.031013
4.801014
7.121015
5.731014
4.321014
4.121015
3.831016
1.621015
3.841014
1.661015
9.001016
1.501015
4.331014
1.961015
5.331014
1.431015
2.351014
2.421014
5.521014
3.371014
1.161015
4.081014
1.031014
6.071014
1.831014
8.491014
5.391014
9.941015
5.991014
3.391014
1.191014
1.041014
8.751014
1.671014
3.991014
7.251014
2.741014
3.181014
8.231015
4.971017
7.511014
4.331014
3.271014
4.141016
9.971016
7.021014

Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]
Radionuclide
Ir-187
Re-187
W-187
Ir-188
Pt-188
Re-188m
Re-188
W-188
Ir-189
Os-189m
Pt-189
Re-189
Ir-190n
Ir-190m
Ir-190
Os-190m
Ir-191m
Os-191m
Os-191
Pt-191
Ir-192m
Ir-192
Au-193
Hg-193m
Hg-193
Os-193
Pt-193m
Pt-193
Au-194
Hg-194
Ir-194m
Ir-194
Os-194
Tl-194m
Tl-194
Au-195m
Au-195
Hg-195m
Hg-195
Ir-195m
Ir-195
Pb-195m
Pt-195m
Tl-195
Hg-197m
Hg-197
Pt-197m
Pt-197
Landolt-Brnstein
New Series VIII/4

6 External dosimetry
Skin Dose
[Sv/(Bq s m2)]
2.031014
0.00
4.091014
9.181014
1.181014
3.911015
5.351014
2.911015
4.141015
7.161018
1.821014
2.151014
8.891014
7.521018
8.241014
9.121014
4.071015
3.671016
4.351015
1.711014
8.811015
5.531014
9.161015
6.211014
1.261014
2.441014
3.071015
2.071017
6.191014
2.651017
1.341013
5.851014
5.221017
1.471013
4.411014
1.351014
4.121015
1.381014
1.111014
3.531014
2.191014
9.971014
5.921015
7.521014
1.021014
3.351015
1.861014
1.061014

Effective Dose
[Sv/(Bq s m2)]
1.541014
0.00
2.131014
7.521014
7.901015
2.561015
3.131015
1.101016
2.771015
1.241019
1.341014
3.081015
6.811014
1.381019
6.321014
7.031014
2.621015
2.311016
2.781015
1.211014
6.841015
3.611014
6.031015
4.691014
7.701015
3.291015
3.761016
4.071019
4.941014
6.231019
1.041013
4.731015
2.171017
1.031013
3.411014
8.521015
2.731015
8.781015
8.381015
1.781014
2.171015
7.121014
2.441015
5.941014
3.621015
2.261015
3.251015
9.731016

Radionuclide
Tl-197
Au-198m
Au-198
Pb-198
Tl-198m
Tl-198
Au-199
Hg-199m
Pb-199
Pt-199
Tl-199
Au-200m
Au-200
Bi-200
Pb-200
Pt-200
Tl-200
Au-201
Bi-201
Pb-201
Tl-201
Bi-202
Pb-202m
Pb-202
Tl-202
Bi-203
Hg-203
Pb-203
Po-203
Tl-204
Bi-205
Pb-205
Po-205
Bi-206
Tl-206
At-207
Bi-207
Po-207
Tl-207
Tl-208
Pb-209
Tl-209
Bi-210m
Bi-210
Pb-210
Po-210
At-211
Bi-211

6-39
Skin Dose
[Sv/(Bq s m2)]
2.431014
3.751014
4.081014
2.661014
7.401014
1.161013
8.231015
2.711014
8.551014
4.381014
1.491014
1.271013
6.361014
1.431013
1.311014
1.131014
7.501014
2.781014
8.991014
4.431014
4.891015
1.571013
1.171013
2.721017
2.631014
1.391013
1.561014
1.871014
1.001013
1.241014
9.701014
2.921017
9.101014
1.901013
3.361014
7.761014
9.311014
7.671014
3.061014
2.341013
9.351015
1.591013
1.631014
2.301014
1.281016
4.811019
1.961015
3.071015

Effective Dose
[Sv/(Bq s m2)]
1.781014
2.391014
1.811014
1.861014
5.261014
9.471014
3.671015
7.631015
6.831014
9.321015
1.021014
9.321014
1.321014
1.081013
8.171015
2.331015
5.981014
2.621015
6.081014
3.351014
3.251015
1.241013
9.291014
4.961019
2.001014
1.131013
1.041014
1.301014
7.591014
1.711016
7.981014
5.451019
7.291014
1.511013
3.951016
6.091014
7.041014
6.081014
4.531016
1.691013
1.001016
9.651014
1.121014
2.581016
4.481017
3.891019
1.371015
2.041015

6-40
Radionuclide
Pb-211
Po-211
Bi-212
Pb-212
Po-212
Bi-213
Po-213
Bi-214
Pb-214
Po-214
At-215
Po-215
At-216
Po-216
At-217
At-218
Po-218
Rn-218
Fr-219
Rn-219
Fr-220
Rn-220
Fr-221
Fr-222
Ra-222
Rn-222
Ac-223
Fr-223
Ra-223
Ac-224
Ra-224
Ac-225
Ra-225
Ac-226
Ra-226
Th-226
Ac-227
Pa-227
Ra-227
Th-227
Ac-228
Pa-228
Ra-228
Th-228
Th-229
Pa-230
Th-230
U-230
Pa-231

6 External dosimetry
Skin Dose
[Sv/(Bq s m2)]
3.061014
4.471016
4.051014
1.351014
0.00
3.391014
0.00
1.281013
2.771014
4.711018
1.121017
1.011017
8.031017
9.571019
1.861017
2.121016
7.561019
4.301017
2.041016
3.381015
8.531016
2.201017
2.021015
4.761014
5.511016
2.281017
3.051016
2.301014
8.871015
1.081014
6.351016
9.401016
3.011015
2.151014
4.791016
6.371016
1.101017
1.081015
3.191014
6.501015
7.881014
6.561014
0.00
1.501016
5.411015
3.731014
4.511017
1.071016
2.441015

Effective Dose
[Sv/(Bq s m2)]
2.591015
3.561016
8.951015
6.241015
0.00
6.161015
0.00
7.251014
1.091014
3.811018
8.511018
7.791018
5.381017
7.751019
1.371017
9.711017
4.211019
3.401017
1.531016
2.461015
4.401016
1.721017
1.321015
5.791016
4.031016
1.771017
1.871016
2.201015
5.471015
8.011015
4.291016
6.371016
2.401016
5.571015
2.841016
3.211016
5.121018
7.381016
7.011015
4.431015
4.491014
5.161014
0.00
8.101017
3.361015
2.911014
1.481017
4.561017
1.571015

Radionuclide
Th-231
U-231
Np-232
Pa-232
Th-232
U-232
Np-233
Pa-233
U-233
Np-234
Pa-234m
Pa-234
Pu-234
Th-234
U-234
Np-235
Pu-235
U-235
Np-236a
Np-236b
Pu-236
U-236
Am-237
Np-237
Pu-237
U-237
Am-238
Cm-238
Np-238
Pu-238
U-238
Am-239
Np-239
Pu-239
U-239
Am-240
Cm-240
Np-240m
Np-240
Pu-240
U-240
Am-241
Cm-241
Pu-241
Am-242m
Am-242
Cm-242
Pu-242

[Ref. p. 6-42
Skin Dose
[Sv/(Bq s m2)]
2.521015
3.821015
6.941014
5.571014
3.441017
5.921017
4.781015
1.661014
4.571017
8.411014
5.481014
1.241013
3.461015
7.501016
4.251017
1.821016
4.781015
8.641015
9.171015
5.761015
4.831017
3.571017
2.141014
1.541015
2.541015
9.971015
5.091014
3.941015
4.311014
4.091017
2.911017
1.561014
1.601014
1.861017
2.611014
5.791014
4.681017
5.931014
9.151014
3.921017
3.121015
1.281015
3.141014
1.171019
1.361016
8.201015
4.291017
3.271017

Effective Dose
[Sv/(Bq s m2)]
4.581016
2.561015
5.381014
4.261014
7.241018
1.171017
3.391015
8.551015
1.421017
6.831014
1.211015
8.721014
2.491015
2.941016
6.111018
4.191017
3.451015
6.461015
4.741015
1.921015
4.681018
3.861018
1.551014
8.871016
1.761015
5.291015
4.041014
2.851015
2.561014
3.501018
2.501018
9.261015
6.951015
3.481018
2.131015
4.671014
4.171018
1.551014
5.881014
3.421018
5.871017
6.741016
2.111014
6.331020
2.491017
6.091016
4.021018
2.901018

Landolt-Brnstein
New Series VIII/4

Ref. p. 6-42]
Radionuclide
Am-243
Cm-243
Pu-243
Am-244m
Am-244
Cf-244
Cm-244
Pu-244
Am-245
Bk-245
Cm-245
Pu-245
Am-246m
Am-246
Bk-246
Cf-246
Cm-246
Pu-246
Bk-247
Cm-247
Cf-248
Cm-248

Landolt-Brnstein
New Series VIII/4

6 External dosimetry
Skin Dose
[Sv/(Bq s m2)]
2.751015
9.791015
8.151015
3.111014
5.251014
4.651017
3.911017
2.691017
1.621014
1.581014
5.361015
4.001014
8.561014
6.421014
5.311014
3.351017
3.491017
8.821015
7.431015
1.791014
3.171017
2.671017

Effective Dose
[Sv/(Bq s m2)]
1.851015
5.301015
9.611016
3.631016
3.591014
4.741018
3.401018
2.081018
1.451015
9.261015
3.491015
1.861014
4.741014
3.061014
4.271014
3.921018
3.101018
5.351015
4.201015
1.381014
3.251018
2.351018

Radionuclide
Bk-249
Cf-249
Cm-249
Bk-250
Cf-250
Cm-250
Es-250
Cf-251
Es-251
Cf-252
Fm-252
Cf-253
Es-253
Fm-253
Cf-254
Es-254m
Es-254
Fm-254
Fm-255
Fm-257
Md-257
Md-258

6-41
Skin Dose
[Sv/(Bq s m2)]
4.071017
1.911014
1.591014
6.431014
3.021017
0.00
2.211014
1.121014
5.351015
3.081017
2.951017
1.661015
4.551017
4.551015
9.831020
3.761014
5.651016
3.431017
3.951016
7.181015
6.201015
1.821016

Effective Dose
[Sv/(Bq s m2)]
4.681019
1.451014
1.021015
4.121014
3.091018
0.00
1.761014
5.011015
3.651015
3.631018
3.451018
1.751017
1.601017
3.121015
1.011020
2.111014
1.571016
4.761018
8.821017
4.151015
4.521015
3.891017

6-42

6 External dosimetry

6.7 References
68Dre
69Sny
69Spi
70Sto
75Emm
75ICR
77ICR
78Sny
79Lic
80Cri
82Kra
83ICR
83Rou
85ICR
85Nel
85Sai
86Gro
88DOE
88ICR
88Zan
89Vei
90Hol
90Sai
91Bri

Drexler, G.: Proceedings of the Symposium on Microdosimetry, Ispra (Italy), 13-15


November 1967. European Communities, Brussels. Report No. EUR 3747 d-f-e, 1968, p. 433.
Snyder, W.S., Ford, M.R., Warner, G.G., Fisher jr., H.L.: Medical Internal Radiation Dose
Committee (MIRD) Pamphlet No. 5, Supplement No. 3: J. Nucl. Med. 10 (1969).
Spiers, F.W.: Delayed effects of bone-seeking radionuclides, Mays, C.W., Jee, W.S.S.,
Lloyd, R.D., Stover, B.J., Dougherty, J.H., Taylor, G. (eds.), Salt Lake City: University of
Utah Press, 1969, p. 95.
Storm, E., Israel, H.I.: Nucl. Data Tables A 7 (1970) 565.
Emmett, M.B.: Oak Ridge National Laboratory Report No ORNL4972. Oak Ridge, TN,
1975.
International Commission on Radiological Protection: ICRP Publication 23. Oxford, UK:
Pergamon Press, 1975.
International Commission on Radiological Protection: ICRP Publication 26. Oxford, UK:
Pergamon Press, 1977.
Snyder, W.S., Ford, M.R., Warner, G.G., Fisher jr., H.L.: Medical Internal Radiation Dose
Committee (MIRD) Pamphlet No. 5, New York: The Society of Nuclear Medicine, 1978.
Lichtenstein, H., Cohen, M.O., Steinber, H.A., Trubetzkoys, E.S., Beer, M.: Computer code
manual of the electric power research institutes (MAGI) EPRI-CCM-8. Palo Alto, California:
Mathematical application group Inc., 1979.
Cristy, M.: Oak Ridge National Laboratory Report No. ORNL/NUREG/TM-367. Oak Ridge,
TN, 1980.
Kramer, R., Zankl, M., Williams, G., Drexler, G.: GSF-Report S-885. Neuherberg, Germany:
GSF-National Research Center for Environment and Health, 1982.
International Commission on Radiological Protection: ICRP Publication 38. Oxford, UK:
Pergamon Press, 1983.
Roussin, R.W., Knight, J.R., Hubbell, J.H., Howerton, R.J.: Report No. ORNL-RSIC-46
(ENDF-335), Oak Ridge, TN: Radiation Shielding Information Center, Oak Ridge National
Laboratory, 1983.
International Commission on Radiation Units and Measurements: ICRU Report 39.
Bethesda, MD: ICRU Publications, 1985.
Nelson, W.R., Hirayama, H., Rogers, D.W.O.: SLAC-265-UC-32. Stanford, CA: Stanford
Linear Accelerator Center, 1985.
Saito, K., Moriuchi, S.: Radiat. Prot. Dosim. 12 (1985) 21.
Grosswendt, B, Roos, M.: Medizin Physik (1986) 265.
Department of Energy DOE/EH-0070 DOE, Washington DC: Department of Energy, 1988.
International Commission on Radiation Units and Measurements: ICRU Report 43.
Bethesda, MD: ICRU Publications, 1988.
Zankl, M., Veit, R., Williams, G., Schneider, K., Fendel, H., Petoussi, N., Drexler, G.:
Radiat. Environ. Biophys. 27 (1988) 153.
Veit, R., Zankl, M., Petoussi, N., Mannweiler, E., Williams, G., Drexler, G.: GSF-Report
3/89. Neuherberg, Germany: GSF - National Research Center for Environment and Health,
1989.
Hollnagel, R.A.: Radiat Prot. Dosim. 30 (1990) 149.
Saito, K., Petoussi, N., Zankl, M., Veit, R., Jacob, P., Drexler, G.: GSF-Report 2/90.
Neuherberg, Germany: GSF-National Research Center for Environment and Health, 1990.
Briesmeister, J.F.: Los Alamos National Laboratory Report LA-12625-M. Los Alamos, New
Mexico, 1977.

Landolt-Brnstein
New Series VIII/4

6 External dosimetry
91Gro
91ICR
91Pet
92Hal
92ICR1
92ICR2
92Zan
93Eck1
93Eck2
93Gro
93ICR
93Yam
94Gro
94Gua1
94Gua2
94Hir
94ICR
94Yam
94Zub
95ISO
95Sai
95Til
96Cha
96Dim
96ICR
97Zan
98Cla
98ICR
98Sai
99Sta
99Zan
00Pel

6-43

Grosswendt, B.: Radiat Prot. Dosim. 35 (1991) 221.


International Commission on Radiological Protection: ICRP Publication 60. Oxford, UK:
Pergamon Press, 1991.
Petoussi, N., Jacob, P., Zankl, M., Saito, K.: Radiat. Prot. Dosim. 37 (1991) 31.
Halbleib, J.A., Kensek R.P., Mehlhorn, T.A., Valdez, G.D., Seltzer, S.M., Berger, M.J.:
Report No SAND-1634 / UC-405. Albuquerque, New Mexico and Livermore, California:
SANDIA National Laboratories, 1992.
International Commission on Radiation Units and Measurements: ICRU Report 47.
Bethesda, MD: ICRU Publications, 1992.
International Commission on Radiation Units and Measurements: ICRU Report 48.
Bethesda, MD: ICRU Publications, 1992.
Zankl, M., Petoussi, N., Drexler, G.: Health Phys. 62 (1992) 395.
Eckerman, K.F., Ryman, J.C.: Federal Guidance Report No. 12. Oak Ridge, TN: Oak Ridge
National Laboratory, 1993.
Eckerman, K.F., Westfall, R.J., Ryman, J.C., Cristy, M.: Oak Ridge National Laboratory
Report No ORNL/TM-12350. Oak Ridge, TN: Oak Ridge National Laboratory, 1993.
Grosswendt, B.: PTB-Report Dos-22, Braunschweig: Physikalisch-Technische Bundesanstalt, 1993.
International Commission on Radiation Units and Measurements: ICRU Report 51.
Bethesda, MD: ICRU Publications, 1993.
Yamaguchi, Y.: Radioisotopes 42 (1993) 35.
Grosswendt, B.: Radiat Prot. Dosim. 54 (1994) 85.
Guaraldi , R., Padoani, F.: ENEA Report RT/ERG/94/17, 1994.
Guaraldi , R., Padoani, F.: ENEA Report RT/ERG/94/21, 1994.
Hirayama, H.: Radiat. Prot. Dosim. 51 (1994) 107.
International Commission on Radiation Units and Measurements: ICRU Report 53.
Bethesda, MD: ICRU Publications, 1994.
Yamaguchi, Y.: Radiat. Prot. Dosim. 55 (1994) 123.
Zubal, I.G., Harrell, C.R., Smith, E.O., Rattner, Z., Gindi, G., Hoffer, P.B.: Med. Phys. 21
(1994) 299.
International Organization for Standardization: ISO FDIS 4037-3, 1995.
Saito, K., Jacob, P.: Radiat. Prot. Dosim. 58 (1) (1995) 29.
Till, E., Zankl, M., Drexler G.: GSF-Report 27/95. Neuherberg, Germany: GSF-National
Research Center for Environment and Health, 1995.
Chartier, J.-L., Grosswendt, B., Gualdrini, G.F., Hirayama, H., Ma, C.-M., Padoani, F.,
Petoussi, N., Seltzer, S.M., Terrisol, M.: Radiat. Prot. Dosim. 63 (1996) 7.
Dimbylow, P.J.: Proc. Voxel phantom development 6-7 July 1996, Dimbylow, P.J. (ed.),
Chilton, UK: National Radiological Protection Board, 1996, p. 1.
International Commission on Radiological Protection: ICRP Publication 74. Oxford, UK:
Pergamon Press, 1996.
Zankl, M., Drexler, G., Petoussi-Henss, N., Saito, K.: GSF-Report 8/97. Neuherberg,
Germany: GSF - National Research Center for Environment and Health, 1997.
Clark, M.J., Chartier, J.-L., Siebert, B.R.L., Zankl, M.: Radiat. Prot. Dosim. 78 (1998) 91.
International Commission on Radiation Units and Measurements: ICRU Report 57.
Bethesda, MD: ICRU Publications, 1998.
Saito, K., Petoussi-Henss, N., Zankl, M.: Health Phys. 74 (6) (1998) 698.
Stabin, M.G., Tagesson, M., Thomas, S.R., Ljungberg, M., Strand, S.E.: Appl. Radiat. Isot.
50 (1999) 73.
Zankl, M.: Health Phys. 76 (1999) 162.
Pellicioni, M.: Radiat. Prot. Dosim. 88 (2002) 279.

Landolt-Brnstein
New Series VIII/4

6-44
00Xu
01Zan
02Eck
02Pet
02Roe

6 External dosimetry
Xu, X.G, Chao, T. C., Bozkurt A.: Health Phys. 78 (2000) 476.
Zankl, M., Wittmann, A.: Radiat. Environ. Biophys. 40 (2001) 153.
Eckerman, K.F. Private communication, 2002.
Petoussi-Henss, N., Zankl, M., Fill, U., Regulla, D.: Phys. Med. Biol. 47 (2002) 89.
Roesler, S., Heinrich, W., Schraube, H.: Radiat. Prot. Dosim. 98 (2002) 367.

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-1

7 Internal dosimetry of radionuclides

This Chapter reviews the behaviour of radionuclides in the body. It summarises the biokinetic and
dosimetric models that have been developed by the International Commission on Radiological Protection
(ICRP) for assessing radiation doses, and hence risks, from intakes by different routes, including
inhalation and ingestion. These models have been widely accepted around the world for use in
radiological protection. They have been incorporated in the European and International Basic Safety
Standards as well as in many national regulations and guidance notes around the world. Future
developments in this area are also examined. Finally, methods that can be used to assess intakes of
radionuclides by direct and indirect monitoring procedures and requirements for dose assessment are
summarised.

7.1 Introduction
People may be exposed to radionuclides in a number of ways. They may be taken into the body as a result
of occupational exposure or uptake from the environment. They are used extensively in medical diagnosis
and treatment as well as in biomedical research. People may also be exposed externally by submersion in
a radioactive cloud.
For occupational exposure, the main route of intake is by inhalation, although a fraction of any
material deposited in the respiratory system will be transferred to the throat and swallowed, giving the
opportunity for absorption in the gastrointestinal tract. Intakes by direct ingestion may occur and some
radionuclides may be absorbed through the intact skin. Damage to the intact skin by cuts or other wounds
can also result in the entry of radionuclides into the body.
For members of the public, the main route of intake of radionuclides will be by ingestion in food and
drinking water although intakes by inhalation may also occur, in particular in the case of accidental
releases into the environment. For medical applications the method of administration will depend upon
the specific nature of the diagnostic investigation or treatment.
Knowledge of the behaviour of radionuclides in the body is important for assessing radiation doses
resulting from intakes or superficial contamination. For occupational and public exposure the calculation
of radiation doses provides the basis for controlling exposures to within accepted limits, for assessing the
consequences of the presence of radionuclides in the working or natural environment or determining the
need for treatment in the case of accidental intakes. In medical situations radiation doses are needed for
optimising diagnostic and treatment schedules. In the case of administration of radionuclides for clinical
research, for example on the behaviour of radiopharmaceuticals, assessment of radiation doses is needed
for estimating risks for ethical considerations.
The radiation dose received by a tissue as a result of the intake of a radionuclide will depend upon a
number of factors. These include: the route of intake, the physico-chemical form, its biokinetic behaviour
and pathways in the body, organ(s) of accumulation, rate of removal (by physical decay, biological
turnover and excretion) and the quality of the emitted radiation (, , ). Biological variation (age, sex,
dietary habits etc) will also influence behaviour in the body. Thus the determination of the radiation dose
to tissues and the assessment of the possible biological effects resulting from the intake of a particular
radionuclide requires a knowledge of all the pertinent physical, physiological, biokinetic and chemical
data.
Landolt-Brnstein
New Series VIII/4

7-2

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

This Chapter reviews the biokinetic behaviour of radionuclides in the body and illustrates the key
features through examples. It summarises the models that have been developed by the International
Commission on Radiological Protection (ICRP) for assessing doses and hence risks from intakes of
radionuclides and examines some future developments in this area. These models have been widely
accepted around the world for use in radiological protection. They provide the basis for dose coefficients
(doses per unit intakes, Sv Bq1) for assessing radiation doses from intakes by inhalation and ingestion
and have been incorporated in the European and International Basic Safety Standards [96E1, 96I2] as
well as in many national regulations and guidance notes around the world. The use of these dose
coefficients for assessing the risks from intakes of radionuclides is illustrated.
A recent development by ICRP has been the issue of a report giving dose coefficients for the embryo
and foetus following intakes of radionuclides by the mother before or during pregnancy [01I1]. A further
report is presently being prepared that will give doses to the newborn child from radionuclides consumed
in mothers milk. The development of the document is summarised. A new dosimetric model for the
human alimentary tract is also being prepared. The conceptual basis for this model is reviewed. Finally,
methods that can be used to assess intakes of radionuclides by monitoring procedures and the
requirements for dose assessment are described in principal; more detailed information is given in
Sections 10.3.2 and 10.3.3 of Chapter 10.

7.2 Biokinetics of radionuclides in the body


The principal routes by which radionuclides may enter and move around the body and which must be
considered in internal dosimetry are summarised in Figure 7.1. Radionuclides passing through the
gastrointestinal tract, or deposited in the air passages of the lungs, in a wound or on the outer layer of skin
will irradiate these tissues. Soluble forms of radionuclide(s) that are transportable can readily enter the
bloodstream and their subsequent fate depends upon their chemical characteristics. If poorly transportable
they will only slowly enter the bloodstream or the lymphatic system. Any insoluble particles entering the
systemic circulation will be taken up by the reticuloendothelial cells of the liver, spleen and red bone
marrow. Here they may remain for up to the life-span of the individual.
To facilitate calculation of doses to tissues following intakes of radionuclides, the ICRP has developed
a number of generalised biokinetic models to describe their movement and behaviour in the body.
Specific models were given by ICRP for adult workers in Publication 30 [79I1, 80I1, 80I2, 88I2] to
describe the behaviour of radionuclides in the main organs of intake the lungs and gastrointestinal tract
as well as the skin. For radionuclides that have entered the blood and systemic circulation, activity
subsequently deposited in tissues was generally assumed to be uniformly distributed throughout them and
therefore the radiation dose depends solely on the organ mass and both the physical half-life and the
biological half-time of the radionuclide (see Chapters 3 and 4). A specific model was needed for the
skeleton, however, because of the morphology of skeletal bone and the heterogeneous distribution of
deposited activity. Biokinetic models were also given in the various parts and supplements of Publication
30 to describe the behaviour of radionuclides in the body after their entry into the blood.
More recently, ICRP has provided age-dependent biokinetic models for selected radionuclides in
Publications 56, 67, 69, 71 and 72 [89I1, 93I1, 95I1, 95I2, 96I1] and has given dose coefficients (Sv
Bq1) for six ages: 3-month-old infants, 1-, 5-, 10- and 15-year old children and adults. The requirement
for age-dependent models and dose coefficients became apparent in the aftermath of the Chernobyl
accident when it was realised that, whilst some countries had developed such models there were no
models that were generally accepted around the world. Such models are essential for assessing doses to
the public from intakes of radionuclides in foods and drinking water, for making comparisons with dose
limits and for informing decisions on the acceptability for consumption of foods that may be marketed in
many countries. For the development of age-dependent models there was a need to include anatomical
and physiological information, such as age dependent mass and turnover rate of the skeleton. These, socalled physiologically based models provide a framework in which both human and animal data on the
behaviour of radionuclides in the body can be integrated and allow a more realistic approach to the
calculation of doses to individuals of different ages. They also have the important advantage that they can
take into account excretion and are therefore more appropriate for the interpretation of bioassay data.
Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-3

Extrinsic removal
Inhalation
Skin
Lymph nodes

Exhalation

Ingestion

Respiratory
tract

Direct absorption

Liver

Gastrointestinal
tract

Transfer
compartment

Sweat

Wound

Subcutaneous
tissue

Other organs

Kidney

Urinary
bladder
Skin
Urine

Faeces

Fig. 7.1. Summary of the main


routes in intake, transfers and
excretion of radionuclides in the
body; [97I2].

In addition to the development of age-dependent biokinetic models a new human respiratory tract
model (HRTM) was issued in Publication 66 [94I2]. This model has been applied in all the recent
calculations of dose coefficients for workers and the public issued by ICRP (see compilations in 94I1,
96I1, 99I1). Table 7.1 summarises the recent ICRP publications giving revised biokinetic models and
dose coefficients.
A further development has been the issue of a report giving dose coefficients for the embryo and
foetus following intakes of radionuclides by the mother before or during pregnancy. ICRP Publication 88
gives biokinetic models for 31 elements and also dose coefficients for selected radionuclides [01I1].
Presently being prepared by ICRP is a further report that will give doses to the newborn child from
radionuclides consumed in mothers milk. The development of the document is summarised in Section
7.2.8. A new dosimetric model for the human alimentary tract is also being developed that will also be
age-dependent. The conceptual basis for this new model is summarised in Section 7.2.2.

7.2.1 Inhalation
A model for describing the deposition and clearance of inhaled radionuclides in adults who are
occupationally exposed was first given in Publication 30 of ICRP [79I1]. This lung model separated the
respiratory system into three distinct regions, the naso-pharynx (NP), the tracheo-bronchial region (TB)
and the pulmonary region (P). It gave information on the deposition of inhaled radionuclides in these
regions as a function of the activity median aerodynamic diameter (AMAD) of inhaled particulates and on
the rate of clearance of the material from the respiratory system in terms of three default clearance
Classes. These had clearance times from the pulmonary part of the respiratory system of days (Class D),
weeks (Class W) and years (Class Y). The default particle size was taken to have an AMAD of 1 m.
Information was also given on the transfer of the three Classes of material to lymphatic tissue associated
with the respiratory system. A main feature of this lung model is that it calculated only the average dose
to the lungs (TB and P regions). Although the model was developed for adults it has been used for
younger ages but without any changes to parameter values, other than organ mass.

Landolt-Brnstein
New Series VIII/4

7-4

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

Table 7.1 Summary of ICRP reports on dose coefficients for workers and members of the public from
intakes of radionuclides.
ICRP
Application Intake
Contents
Publication
No. (year)
56 (1989)
Publica
Inhalation and Age-dependent systemic models, and tissue dose
ingestion
coefficients for selected radioisotopes, for H, C, Sr, Zr, Nb,
Ru, I, Cs, Ce, Pu, Am and Np. Issued before ICRP
Publication 60 [91I1], and hence giving dose equivalents
using the tissue weighting factors from ICRP Publication 26
[77I1]. It was also issued before ICRP Publication 66 [94I2]
and hence used the ICRP Publication 30 lung model [79I1].
The dose coefficients given in ICRP Publication 56 were
superseded by those in ICRP Publications 67 and 71, which
used the tissue weighting factors from Publication 60.
67 (1993)
Publica
Ingestion
Age-dependent systemic models, and tissue dose
coefficients for selected radioisotopes, for S, Co, Ni, Zn,
Mo, Tc, Ag, Te, Ba, Pb, Po and Ra. Updated systemic
models are given for Sr, Pu, Am and Np.
68 (1994)
Workers
Inhalation and Effective dose coefficients for workers, for about 800
ingestion
radionuclides: selected radioisotopes of the 91 elements
covered in ICRP Publication 30, Parts 1-4 [79I1, 80I1 and
I2, 88I2]. The inhalation dose coefficients for workers
exposed to 226Ra given in ICRP Publication 68 were revised
in Annexe B of ICRP Publication 72. Applies the Human
Respiratory Tract Model, HRTM [94I2].
69 (1995)
Publica
Ingestion
Age-dependent systemic models, and tissue dose
coefficients for selected radioisotopes, for Fe, Sb, Se, Th
and U.
71 (1995)
Publica
Inhalation
Tissue dose coefficients for selected radioisotopes of
elements covered in ICRP Publications 56, 67 and 69, plus
Ca and Cm for which age-dependent systemic models are
given. Applies the HRTM [94I2].
72 (1996)
Publica
Inhalation and Effective dose coefficients for members of the public for
ingestion
radioisotopes of the 31 elements covered in ICRP
Publications 56, 67, 69, and 71, plus radioisotopes of the
further 60 elements covered in ICRP Publications 30 and
68. Applies the HRTM [94I2].
CD-ROM
Publica
Inhalation and A database of equivalent doses to individual tissues
(1998)
and
ingestion
corresponding to the effective dose coefficients in ICRP
workers
Publications 68 and 72. Inhalation dose coefficient for
10 particle sizes.
88 (2001)
Embryo
Inhalation and Dose coefficients for the offspring for intakes by the mother
and foetus ingestion by
(worker or public) before or during pregnancy of
the mother
radionuclides of the 31 elements covered in Publications 68
and 72.
CD-ROM2
Embryo
Inhalation and Database of dose coefficients extending information on
(2002)
and foetus ingestion by
radionuclides in Publication 88.
the mother
a Age-dependent dose coefficients (3 months, 1-, 5-, 10-, and 15-years and adult)

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-5

The Human Respiratory Tract Model (HRTM) described in ICRP Publication 66 [94I2] was
developed to replace the lung model given in ICRP Publication 30. It takes into account extensive data on
the behaviour of inhaled materials that had become available since the ICRP Publication 30 model was
developed. As in the earlier model, deposition and clearance are treated separately. The scope of the
model was extended to apply explicitly to all members of the population, giving reference values for
3-month-old infants, 1-, 5-, 10- and 15-y-old children and male and female adults. The main features of
the model are summarised below.
In the new model, the respiratory tract is represented by five regions (Fig. 7.2). The extrathoracic (ET)
airways are divided into ET1, the anterior nasal passage and ET2, which consists of the posterior nasal and
oral passages, the pharynx and larynx. The thoracic regions are bronchial (BB: trachea, generation 0 and
bronchi, airway generations 1-8), bronchiolar (bb: airway generations 9-15), and alveolar-interstitial (AI:
the gas exchange region). Lymphatic tissue is associated with the extrathoracic and thoracic airways
(LNET and LNTH, respectively). Reference values of dimensions and scaling factors for subjects of
different ages are specified in the model. A main feature of the HRTM, compared with the Publication 30
model, is the calculation of doses to these specific tissues in the five regions and allowance for their
differences in radiosensitivity.

Posterior
Nasal Passage

Pharynx

ET 1
Extrathoracic

Nasal Part
Oral Part

ET 2

Larynx

BB
Trachea
Thoracic
Bronchial
Main Bronchi
Bronchi

Bronchioles

bb

Bronchiolar
Alveolar
Interstitial

Al

bb
Bronchioles
Terminal Bronchioles

Al

Respiratory Bronchioles
Alveolar Duct + Alveoli

Landolt-Brnstein
New Series VIII/4

Fig. 7.2. Respiratory tract regions defined in the Human


Respiratory Tract Model; [94I2].

7-6

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

7.2.1.1 Deposition
The amount of an aerosol inhaled depends upon breathing parameters and these are influenced by age,
body size and level of physical exertion. Deposition of inhaled particles in the HRTM is calculated for
each region of the respiratory system, with account taken of both inhalation and exhalation. This is done
as a function of particle size, breathing parameters and/or work load, and is assumed to be independent of
chemical form. Four standard levels of activity are defined in the HRTM ranging from sleep through to
heavy exercise, and the different proportions of time spent at these reference levels are specified for
representative individuals at the six standard ages [94I2]. The fraction of inhaled aerosol deposited in the
various regions of the lung depends in turn upon the particle size, typically taken as a log-normal
distribution. Age dependent default deposition parameters are given for a range of particle sizes from
0.6 nm activity median thermodynamic diameter (AMTD) to 20 m activity median aerodynamic
diameter (AMAD). Previously, a 1 m AMAD was taken as the default particle size for occupational
exposure, but ICRP Publication 66 now recommends 5 m AMAD as being more typical of the
workplace. For members of the public the default is taken as 1 m AMAD. Table 7.2 compares regional
deposition in the respiratory system for the models of ICRP Publications 30 and 66. For the old and new
defaults for workers, total deposition is about 30 % higher in the new model (82 % c.f. 63 %), with the
extrathoracic region dominating, although a large fraction of this is in the ET1 region and thus unavailable
for systemic uptake. Conversely, deposition in the deep lung (bronchiolar and alveolar-interstitial regions)
is a factor of four higher in the ICRP 30 model (25 % c.f. 6.4 %).
Table 7.2 Comparison of regional deposition for ICRP 30 Lung and ICRP 66 Respiratory Tract Models.
Publication 30 model
Publication 66 model
Adult
Adult mem- Worker
ber of public
1 ma
1 ma
1 ma
5 ma
Region
[%]
Region
[%]
[%]
[%]
Nasal passage (NP)
30
Extrathoracic (ET1)
15
17
34
Extrathoracic (ET2)
19
21
40
Total
(34)
(38)
(74)
Trachea and bronchial tree
(TB)

Bronchial (BB)
Bronchiolar (bb)
Total

1.3
2.0
(3.3)

1.2
1.7
(2.9)

1.8
1.1
(2.9)

Pulmonary region (P)

25

Alveolar-interstitial
(AI)

11

11

5.3

Total

63

Total

48

52

82

a Activity Median Aerodynamic Diameter (AMAD)


The variation in deposition parameters between individuals, depending upon age, gender and habits, is
an important difference from the Publication 30 model [79I1] for which particle size was the only factor
that influenced deposition (see 1 m entries of Table 7.2). Previously, this distinction was not needed,
since the Publication 30 model was intended only for reference adults who were occupationally exposed.
In contrast, the ICRP Publication 66 model [94I2] has been designed for application to all members of the
population. In the new model, deposition, but not clearance, is strongly influenced by age.

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-7

7.2.1.2 Clearance
Subsequent to deposition, material is cleared from the respiratory tract. For material deposited in the
anterior nose ET1, clearance is affected extrinsically by such means as nose-blowing or sneezing. The ET1
deposit is cleared directly from the body and makes no subsequent contribution to gut or systemic tissue
doses. Removal from all other regions is treated as two competing processes: particle transport (by
mucociliary clearance to the throat or translocation to lymph nodes) and absorption to blood.
It is assumed that these clearance processes compete independently with each other and have no age
or gender dependence. Transport processes include mechanical transport to the gut by mucociliary action
and removal by macrophages to the lymph nodes. Particle transport rates are taken to be fixed for all
materials and a single compartment model describes clearance by this mechanism (Fig. 7.3). Absorption
rates, however, are determined by solubility of inhaled materials and default parameters are recommended
for Fast (Type F), Moderate (Type M) and Slow (Type S) absorption. This corresponds roughly to the
ICRP Publication 30 classification scheme and chemical forms previously assigned to Class D, W or Y
are now provisionally treated as Type F, M or S, respectively. The correspondence between the two
schemes is not exact, e.g. the D, W or Y classification refers to whether total pulmonary lung clearance
(by absorption to blood or clearance to the throat and then through the gut) is of the order of days, weeks
or years, whereas Type F, M or S refers only to the absorption component. The mechanical clearance of
the deposited activity is not dependent on the chemical form. The main clearance components for the two
models, in the form of approximate biological half-times, are summarised in Table 7.3. Qualitatively,
residence times in the lung are reduced, quite drastically, for Type F compared to Class D and elevated
for Types M and S relative to Classes W and Y.
Anterior
nasal
Nasooropharynx/
larynx

Extrathoracic
LNET

LNTH

ET2

ETseq

0.01

Bronchi

Bronchioles

ET1

0.001

0.01

BB2

bb2

GI tract

BB1
2

0.03
bbseq

100

Environment

10

0.03
BBseq

bb1
0.0001 0.001 0.02

Alveolar
interstitial

0.00002

AI3 AI2 AI1

Thoracic

Fig. 7.3 Compartment model representing time-dependent particle transport from


each respiratory tract region. Rates shown alongside arrows are reference values in
units of d1. It is assumed that (i) the AI deposit is divided between AI1, AI2 and
AI3 in the ratio 0.3:0.6:0.1; (ii) the fraction of the deposit in BB and bb that is
cleared slowly (BB2 and bb2) is 50 % for particles of physical size <2.5 m and
decreases with diameter >2.5 m, and the fraction retained in the airway wall
(BBseq and bbseq) is 0.7 % at all sizes; (iii) 0.05 % of material deposited in region
ET2 is retained in its wall (ETseq) and the rest in compartment ET2 which clears
rapidly to the GI tract. The model as shown above would describe the retention and
clearance of a completely insoluble material. However, there is in general
simultaneous absorption to body fluids of material from all the compartments
except ET1; [94I1].

Landolt-Brnstein
New Series VIII/4

7-8

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

Table 7.3. Comparison of approximate clearance half-times for Publication 30 Lung and Publication 66
Respiratory Tract Model
ICRP 30
ICRP 66

Class
D
W
Y

Pulmonary clearance
T
0.5 d (100 %)
50 d (60 %)
500 d (60 %)

Type
F
M
S

Dominant absorption
T
10 min (10 %)
140 d (90 %)
7000 d (99.9 %)

By considering the relative rates of the two independent clearance processes (mechanical and
absorption) in the HRTM and the amount absorbed in the gut after clearance to the throat it is possible to
calculate the fraction of material ultimately transferred to the blood and systemic circulation, both directly
from the lungs and indirectly via the gut. Because particle transport rates are fixed for all lung Types, the
proportion of material escalating to the gut increases as the classification changes from Type F to Type S.
It is interesting to compare the amounts transferred to the circulation for inhalation of the different Types.
This is illustrated in Fig. 7.4, ignoring the effect of radioactive decay. For Type F material, such as
soluble (transportable) forms of radioisotopes of caesium and iodine (e.g. 137Cs, 131I) there is rapid
translocation to the blood with about 25 % of the intakes being taken up by within a day. In contrast for
Type S materials, such as 239PuO2, transfer to the blood is much slower with about 0.15 % transferred after
1000 days. Examples of the lung clearance Types adopted for various chemical forms of a selection of
radionuclides are given in Table 7.4. Although the HRTM model provides these default clearance Types
there is also provision for including material specific absorption parameters when information is
available. ICRP has issued a guidance document on this application of the HRTM [02I1].

Fraction of inhaled activity transferred to blood

1
Type F
10-1

Type M

10-2
Type S
10-3

10-4
10-2

10-1

1
10
10 2
Time after intake [d ]

10 3

10 4

Fig. 7.4. Cumulative fraction of inhaled activity absorbed into blood directly from the respiratory tract as a
function of time after intake for each default absorption
Type (in the absence of radioactive decay), for a reference worker; [02I1].

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-9

Table 7.4 Examples of lung clearance Types adopted by ICRP for workers; [94I1].
Radionuclide
Chemical form
Inhalation Type
Tritium
Tritiated water
SR2a
Cobalt
Unspecified compounds,
Mb
Oxides, hydroxide, halides and nitrate
S
Strontium
Unspecified compounds
Fb
Strontium titanate
S
Zirconium
Unspecified compounds
F
Oxides, hydroxide, halides and nitrate
M
Zirconium carbide
S
Niobium
Unspecified compounds
M
Oxides and hydroxide
S
Ruthenium
Unspecified compounds
F
Halides
Mb
Oxides and hydroxides
S
Iodine
All compounds
Fb
Vapour
Vb
Caesium
All compounds
Fb
Cerium
Unspecified compounds
Mb
Oxides, hydroxides and fluorides
S
Polonium
Unspecified compounds
F
Oxides, hydroxides and nitrate
M
Radium
All compounds
M
Thorium
Unspecified compounds
M
Oxide and hydroxides
S
Uranium
Most hexavalent compounds, e.g. UFO6,
F
UO2F2 and UO2(NO3)2
Less soluble compounds, e.g. UO3, UF4,
Mb
UCI4 and most other hexavalent compounds
Highly soluble compounds, e.g. UO2, U3O8
S
Plutonium
Unspecified compounds
Mb
Insoluble oxides
Sb
Americium
All compounds
Mb
Sc
Trace contaminant
Curium
All compounds
Mb
a Excretion and retention functions for inhalation of 3H2O given in Figure 7.15.
b Excretion and retention functions for inhalation of 5 m AMAD aerosols given in
Figures 7.16-7.25.
c Trace contaminant formed from 241Pu in matrices of nuclear fuel in insoluble
(Type S) forms (Fig. 7.25).
7.2.1.3 Gases and vapours
For radionuclides inhaled in particulate form, it is assumed that entry into and deposition in the
respiratory tract is governed by the size distribution of the aerosol particles [94I2]. The situation is
different for gases and vapours, for which the radionuclide has a specific behaviour at its site of entry into
the respiratory tract, depending on the chemistry of the compound. Almost all inhaled molecules contact
airway surfaces, but usually return to the air unless they dissolve in, or react with, the surface lining. The
fraction of an inhaled gas or vapour that is deposited in each region thus depends on its solubility and
reactivity. Generally, however, the regional deposition of a gas or vapour cannot be predicted on a
mechanistic basis, from knowledge of its physical and chemical properties, but has to be obtained from an
in vivo experimental study.
Landolt-Brnstein
New Series VIII/4

7-10

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

The HRTM assigns gases and vapours to three classes:


Class SR-1 (soluble or reactive). Deposition may occur throughout the respiratory tract. Retention in
respiratory tract tissues and uptake to the systemic circulation may be less than 100 % of the inhaled
activity, although this is the default assumption; e.g. tritium gas and tritiated methane, carbon
monoxide.
Class SR-2 (highly soluble or reactive). Total deposition occurs in the extrathoracic airways (ET2).
Subsequent retention in the respiratory tract and absorption to body fluids are determined by the
chemical properties of the specific gas or vapour; e.g. tritiated water, organically bound tritium and
carbon dioxide.
Class SR-0 (insoluble and non-reactive). Negligible deposition in the respiratory tract. External
irradiation from submersion in the cloud of gas, and internal irradiation from gas within the
respiratory tract. e.g. from all radioisotopes of argon, krypton and xenon (except 37Ar, 94I1).
Subsequent retention in the respiratory tract and absorption to body fluids are determined by the
chemical properties of the specific gas or vapour.
ICRP Publications 68 and 71 as well as the guidance document [94I1, 95I2, 02I1] give information on
the assignment of gases and vapours to these three classes, and for selected Class SR-1 compounds
information on fractional deposition and subsequent clearance.
As an alternative to any of the three default Types defined in ICRP Publication 66, very fast uptake to
body fluids (Type V) may be recommended. Although consideration has to be given to the total
respiratory tract deposition, regional deposition does not need to be assessed for such materials, since, for
the purposes of dose calculation, they can be treated as if they were injected directly into body fluids.
Examples are tritiated water and tritiated methane, methyl iodide and methane.
7.2.1.4 Dosimetry
In the ICRP Publication 30 lung model, doses to the respiratory system were averaged over 1 kg of lung
tissue and the energy of charged particle emissions in the TB, P and respiratory lymph node regions was
assumed to be completely absorbed within the lung, i.e. the absorbed fractions were unity for charged
particles. In the HRTM model, doses are calculated to several specific regions of the lung and account is
taken of variations in radiosensitivity. Absorbed fractions are energy dependent and prescribed functions
are given for all source and target combinations and particle types.
The target cells identified for the assessment of doses are: basal cells of the epithelium in both
extrathoracic regions; basal and secretory cells in the bronchial epithelium; Clara cells (a type of secretory
cell) in the bronchiolar epithelium; and endothelial cells, such as those of capillary walls and type II
epithelial cells, in the alveolar-interstitial (AI) region.
The overall dose to the lung is then taken to be a weighted sum of the doses to the following regions:
bronchial, bronchiolar, pulmonary and lymphatic with weighting factors of 0.333, 0.333, 0.333 and 0.001,
(the sum is 1), respectively. These weights are known as regional apportionment factors to distinguish
them from the tissue weighting factors used in the calculation of effective dose (Chapter 4). They
represent the contribution from each region towards the total radiation detriment associated with
irradiation of the lung.

7.2.2 Ingestion
The model of the gastrointestinal tract (GI) presently used by ICRP to describe the behaviour of ingested
radionuclides is that given in ICRP Publication 30 [79I1]. Radionuclides contaminating food or drink, or
cleared from the lung by mucociliary action are swallowed, pass down the oesophagus and enter the
gastrointestinal (GI) tract, which is treated as four compartments (Fig. 7.5). Absorption is usually
described by f1 values which give fractional absorption into the systemic circulation (e.g. f1 = 1.0,
absorption = 100 %, f1 = 0.01, absorption = 1 %). The transport of material through the GI tract is
described in terms of movement through the four regions.
Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-11

Stomach (ST): Its contents are acidic, and little absorption takes place other than for very soluble
radionuclides such as caesium or iodine for which absorption from the stomach is assumed to be complete
(f1 = 1.0). All other radionuclides are assumed to be absorbed in the small intestine. The residence time
for food in the stomach varies from minutes to hours depending on many factors the amount and
composition of food, exercise and emotions. In the dosimetric model the mean residence time is taken to
be 1 h.
Small Intestine (SI): The principle site of absorption. The contents are alkaline, so that elements which
hydrolyse such as rare earths and actinides are not normally readily absorbed. The mean residence time is
assumed to be 4 hours. Recommended f1 values are given in ICRP publications for specific radionuclides
(e.g. 226Ra: f1 = 0.2, 144Ce: f1 = 0.0005, 239PuO2: f1 = 0.00001).
Upper Large Intestine (ULI): Water is absorbed here from the semi-liquid contents. The mean
residence time is taken to be 13 hours.
Lower Large Intestine (LLI): This region acts as a store for food residues and often forms the critical
organ for long-lived non-transportable ingested radionuclides. The mean residence time is taken to be
24 hours.

6WRPDFK 67 

6PDOOLQWHVWLQH 6,

%RG\IOXLGV

8SSHU/DUJH
,QWHVWLQH

/RZHU/DUJH
,QWHVWLQH

Fig. 7.5. Biokinetic model for the gastrointestinal tract (based upon 79I1).

Table 7.5 gives the transit time and mass of the contents for the different regions of the GI tract that
are assumed for dose calculations. Typical f1 values are given in Table 7.6 for some important elements.

Landolt-Brnstein
New Series VIII/4

7-12

7 Internal dosimetry of radionuclides

Table 7.5 Regional masses and residence times in the


gastrointestinal tract; [79I1].
Portion of GI tract that is the
Mass of contents [g]
critical tissue
Stomach (ST)
250
Small Intestine (SI)
400
Upper Large Intestine (ULI)
220
Lower Large Intestine (LLI)
135
a Transfer rate

[Ref. p. 7-68

ICRP Publication 30 dosimetric model for the


Mean residence time [days]

Ka days1

1/24
2/24
13/24
24/24

24
6
1.8
1

Table 7.6 Examples of f1 values adopted by ICRP for workers; [94I1].


Radionuclide
Chemical form
f1
Tritium
Tritiated water
1.0a
Organically bound tritium
1.0a
Cobalt
Unspecified compounds
0.1a
Oxides, hydroxides and inorganic
0.05
compounds
Strontium
Titanate
0.01
Unspecified compounds
0.3a
Zirconium
All compounds
0.002
Niobium
All compounds
0.010
Ruthenium
All compounds
0.05a
Iodine
All compounds
1.0a
Caesium
All compounds
1.0a
Cerium
All compounds
5 104 a
Polonium
All compounds
0.10
Radium
All compounds
0.20
Thorium
Oxide and hydroxides
2.0 104
Unspecified compounds
5.0 104
Uranium
Unspecified compounds
0.02a
Most tetravalent compounds, e.g. UO2,
0.002
U3O8, UF4
Plutonium
Nitrate
1 104 a
Insoluble oxides
1 105 a
Unspecified compounds
5 104 a
Americium
All compounds
5 104 a
Curium
All compounds
5 104
a Chemical forms of radionuclides for which retention and excretion functions
are given in Figures 7.15-7.25.
The f1 values recommended for workers are not necessarily appropriate for food and drinking water.
Moreover the absorption of radionuclides tends to be greater in the newborn although the results of
animal studies suggest that gut absorption decreases as age increases, reaching adult f1 values by about the
time of weaning in most cases. An expert group set up by the Nuclear Energy Agency (NEA) within the
Organisation for Economic Cooperation and Development (OECD) [88N1] suggested f1 values to be used
as average values for the first year of life. The expert group recommended that for fractional absorption
values between 0.01 and 0.5 in adults, an increase by a factor of 2 be assumed for the first year of life; but
for elements with a fractional absorption in adults of 0.001 or less, a value 10 times that of the adult
should be assumed. This general approach has been adopted in the current ICRP documents when more
specific data are not available.

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-13

In the dosimetric model for the GI tract doses are calculated to the radiosensitive mucosal cell layer.
For low LET radiation ( particles and -rays) this is nominally taken to be one half of the average energy
absorbed per gram of the contents and for high LET () radiation one two-hundredth.
The Publication 30 model for the GI tract has a number of distinct limitations in use and although it
has been used for calculating doses to infants and children it does not have specific age-dependent
parameter values. For this reason a Task Group of ICRP is developing a new model for the Human
Alimentary Tract (HAT). The proposed new model is illustrated in Figure 7.6.

Salivary
glands

Oral cavity
Pharynx

Teeth

Oeso Oeso phagus 1 phagus 2

Oral
mucosa

Slow

Fast

B
L
O
O
D

Secretory
organs

Stomach

Stomach
wall

Hepatic
artery

Small intest

SI
wall

Liver

Left colon

LC
wall

Right colon

RC
wall

Sigmoid
Rectum

SR
wall

B
L
O
O
D

Portal vein

Fig. 7.6. Proposed structure for


the new human alimentary tract
model; [03M1].

The revision of the Publication 30 model was motivated by a number of developments:


The 1990 recommendations of ICRP introduced specific risk estimates and tissue weighting factors,
wT for radiation-induced cancer of the oesophagus, stomach and colon, requiring dose estimates for
each of these regions [91I1]. The Publication 30 model did not include the oral cavity, or the
oesophagus and treated the colon as two regions upper and lower large intestine (Figure 7.5). The
new model for the alimentary tract will comprise the oral cavity, including the mouth, teeth, salivary
glands and pharynx, the oesophagus, the stomach, the small intestine, including duodenum, jejunum
and ileum, the large intestine, including ascending, transverse and descending colon, rectum and anal
canal.
Since the development of the ICRP Publication 30 model, a considerable body of data has become
available on the transit of materials through the different regions of the alimentary tract. These data
have been obtained using non-invasive, mainly scintigraphic techniques and include studies of
differences between solid and liquid phases, age and gender related differences and the effect of
disease conditions. These data are being used to set default transit rates for the defined regions
of the alimentary tract for the six age groups given in ICRP Publication 56 (Table 7.1) [89I1].
Information has become available for morphometrical and physiological parameters and on the
location of sensitive cells in different regions of the alimentary tract.
More information has become available concerning absorption, retention and transfer from different
regions of the alimentary tract.
Extensive age-, gender- and health-dependent information is available.

Landolt-Brnstein
New Series VIII/4

7-14

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

Development of the new model has been described [03M1]. The new HAT model will be applicable
to children and adults under all circumstances of exposure. It considers the movement of radionuclides
throughout the alimentary tract from ingestion to elimination. It takes account of sites of radionuclide
absorption and retention in the alimentary tract and routes of secretion of absorbed radionuclides into the
alimentary tract. Doses will be calculated for sensitive cells in each region: mouth, oesophagus, stomach,
small intestine and colon.
The new model is more detailed and morphological than the previous gastro-intestinal tract model
[79I1]. The new model is physiologically based. It includes consideration of absorption in regions other
than the small intestine when such information is available. The model can also be used for radiopharmaceuticals. The model provides the flexibility needed to calculate dose to the alimentary tract for a
wide range of exposure conditions and for specific individuals. A gut transfer factor, equivalent to the f1
value, will take account of absorption from the small intestine and from other regions of the alimentary
tract where information is available.
An important development in the new model is the calculation of doses to sensitive cells in the
different regions of the alimentary tract. The location of sensitive epithelial stem cells in the various
regions is considered separately; that is for the mouth, oesophagus, stomach, small intestine and colon.
Doses from radionuclides in the gut lumen, retained radionuclides and radionuclides in transit to blood
are considered.
It is expected that the report will be used as the basis for future dosimetric calculations for both
ingested radionuclides and radionuclides passed through the throat and swallowed after inhalation.

7.2.3 Cuts and wounds


The presence of cuts, abrasions, burns or other pathological damage to the skin may greatly increase the
ability of radioactive materials to reach subcutaneous tissues and thence the blood and systemic
circulation. Although much of the material deposited at a wound site may be retained at the site, and can
be surgically excised, soluble (transportable) material can be transferred to the blood and hence to other
parts of the body. These events occur only as a result of accidents, each event will, therefore, be unique
and need to be assessed by occupational health physicists and medical staff. To date, ICRP has not given
advice on the interpretation of wound monitoring data following accidents involving radionuclides. The
biokinetic models that have been developed for various radionuclides are, however, applicable to the
soluble component of any deposit in cuts or wounds that enters the blood circulation. Insoluble material
will be slowly translocated to regional lymphatic tissue, where it will gradually dissolve and eventually
enter the blood. A variable fraction of insoluble material can be retained at the wound site or in lymphatic
tissue for the remainder of the individual's life. If particulate material enters the blood it deposits
principally in phagocytic cells in the liver, spleen and bone marrow.
The United States National Committee on Radiological Protection and Measurements (NCRP) has
established a Committee to review the problem of wound dosimetry. The report it is preparing will
contain an extensive compilation of human and experimental data on the behaviour of radionuclides at
wound sites. Four default categories for wound retention of deposited material have been proposed as
summarised below:

weakly retained (10 % retained at one day, <1 % retained at 16 days);


moderately retained (11-55 % retained at one day; 5 % at 64 days);
strongly retained (32-85 % retained at one day; 8-40 % at 64 days); and
avidly retained (>80 % retained in one day; >50 % at 64 days).

In addition default categories are also being considered for colloids, particles and fragments.
In reviewing the experimental data available, various chemical forms of elements/radionuclides are
being allocated to these categories on the basis of either the results of studies in experimental animals or
their chemical characteristics [03G1]. Once the report is complete it will need to be reviewed by the
NCRP and it is possible these categories may change. The ICRP is awaiting publication of the report
before deciding on its future work in this area.
Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-15

7.2.4 Absorption through intact skin


The intact skin provides an effective barrier against the entry of most radioactive materials into the body,
exceptions of practical importance being tritium oxide as liquid or vapour, organic carbon compounds and
iodine as vapour or in solution. No generalised model has been adopted by ICRP for estimating
absorption of radionuclides through the skin although it would be possible to develop specific models.
For example, the behaviour of tritiated organic compounds following direct absorption through the skin
would be expected to be significantly different from that after inhalation or ingestion. For skin
contamination, both the radiation dose to the area of skin contaminated and the dose to the whole body as
a result of absorption need to be considered. ICRP [77I1, 91I1] has recommended that for skin
contamination doses should be calculated to sensitive cells, assumed to be at a depth of 70 m (as a
reasonable average value). ICRP [79I1] addressed the uptake of tritiated water vapour by assuming the
uptake is instantaneously distributed within body water in the same manner as the inhaled water vapour.
That is, for airborne HTO vapour, the dose per unit uptake through the intact skin is the same as the dose
per unit activity inhaled. For deposited activity doses are to be calculated as an average to each cm2 of
skin tissue. This applies to activity distributed over the skin surface or aggregated in particles. No specific
models are recommended by ICRP for calculating doses from particles deposited on the skin (also see
Section 8.2).

7.2.5 Systemic behaviour of radionuclides


The fraction of an intake of a radionuclide entering the systemic circulation is referred to as the uptake. In
Publication 30 [79I1, 80I1, 80I2, 86I1] ICRP reviewed biokinetic data for each element for use in the
calculation of limits on internal exposure to radionuclides by workers for intakes by inhalation and
ingestion. Element-specific biokinetic models were given for the distribution and retention of
radionuclides following their entry into the blood. The ICRP 30 models applied specifically to workers
and not to members of the public. More recently, Publications 56, 67, 69 and 71 have revised the
biokinetic models for selected elements and these have been applied in the calculation of dose coefficients
for both workers and members of the public [89I1, 93I1, 94I1, 95I1, 95I2] (Table 7.1).
If a radionuclide that enters the blood is an isotope of an element that is required by the body then it
will follow the normal metabolic pathways for that element (e.g. Na, P, K, Ca, Fe). If it has similar
chemical properties to an element that is normally present then it will tend to follow the biokinetic
pathways of that element, although its rate of movement between the various compartments in the body
may be different (e.g. 90Sr and 226Ra behave similarly to Ca, 137Cs and 86Rb similarly to K). For other
radionuclides their behaviour in the body will depend upon their affinity for biological ligands and other
transport systems in the body and, as a result, the extent of uptake is unpredictable and must be assessed
from the available human or animal data (e.g. 95Nb, 106Ru, 239Pu, 241Am).
Radionuclides entering the blood may distribute throughout the body (e.g. 3H, 42K, 137Cs); they may
selectively deposit in a particular tissue (e.g. 131I in the thyroid; 90Sr in bone) or they may deposit in
significant quantities in a number of tissues (e.g. 239Pu, 241Am, 144Ce). Some examples of the behaviour of
selected radionuclides are given below. Limited information is also given on methods of treatment for
accidental intakes. More information on decorporation of radionuclides is given in Chapter 9.
7.2.5.1 Elements that distribute widely in body tissues
Hydrogen
Tritium labelled water (HTO), given either orally or by intravenous injection, is rapidly absorbed from the
lungs and absorption from the gut is also essentially complete (f1= 1.0). HTO distributes throughout the
body water and is subsequently lost from the body with a biological half-time of about 10 days as a result
of excretion in the urine, sweat, faeces and via the lungs (i.e. about 7 % of the total body water is lost per
day). The addition of HTO to the body water has been a standard method of determining total body water
by isotope dilution. For example, following intravenous injection of 1000 kBq of HTO the activity in a
urine sample 6 hours later was 20 Bq ml1. The total body water is then:
Landolt-Brnstein
New Series VIII/4

7-16

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

1,000,000 Bq
= 50,000 ml (50 litres)
20 Bq/ml
The total body water in Reference Man is 42 litres [75I1]. The rate of loss of tritiated water can be
increased by increasing the fluid intake (see Chapter 9). Doubling the fluid intake from 2 to about 4 litres
per day can reduce the half-time of tritiated water to about 5-6 days. This rate of loss of tritiated water is
also found in countries with a hot climate, such as India.
In practice, a small fraction of tritium in body tissues becomes incorporated into organic compounds
amino acids, carbohydrates, etc. and is retained with a longer half-time. For adults, ICRP [89I1]
assumes that this fraction is 0.03 (3 %) and is lost with a half-time of 40 days, while the half-time of 10
days applies to the remaining fraction of 0.97 (97 %).
For members of the public, ingesting foods containing tritium, absorption of organically-bound forms
and their incorporation into body tissues will lead to longer retention of a larger component. ICRP [89I1]
assumes that in the adult, the half-times of 10 days and 40 days apply to equal fractions (0.5) of activity
entering blood; values the same as those recommended for organically bound forms of 14C.
Total body water =

Caesium
137

Cs, together with 90Sr, is a major component of nuclear fission. As a result of nuclear weapons testing
Cs has been injected into the atmosphere and weapons fallout has resulted in the contamination of the
food chain and man [77U1]. The first observation of the presence of weapons fallout 137Cs in man was
reported in 1956 [56M1]. Since then it has been shown to be present in everyone as a result of
contamination of the environment by nuclear test explosions, routine releases from nuclear sites and by
the accident at Chernobyl in 1986. 137Cs may enter the body either by inhalation or through the foodchain.
Absorption from the gut is almost 100 % (f1 = 1.0, see Section 7.2.2). Once inside the body caesium ions
(Cs+) behave very similarly to potassium ions (K+) and are rapidly taken up by cells. There is a
considerable concentration factor between the cells and plasma. Generally tissue/plasma concen-tration
ratios are the same for K+ and Cs+ but there are a number of exceptions and some variations between
different species. In particular, muscle accumulates Cs+ more effectively than K+ and is the main site of
long term deposits in the body. At equilibrium the muscle accounts for more than 50 % of the total body
137
Cs in man and bone about 8 %. Accumulation of Cs+ by cells is both by diffusion and by the ion pump
that normally accumulates K+. There is a continual turnover of Cs+ in body tissues.
For the purposes of dosimetry the retention of 137Cs in man is taken to have two components [89I1].
The first accounts for about 10 % of the administered activity and is excreted mainly in the urine with a
half-time(T) of about 2 days. The second component (90 %) has a half-time of about 110 days in males
(range about 50 to 150 days). The long half-time mainly reflects the slow turnover rate in muscle tissue.
The retention half-time for the long-term component in females is less than in males, with a mean value
for adults of about 60 - 65 days. The use of the ICRP value of 110 days is therefore likely to be
conservative for adult females. In children the half time is less than in adults. Thus for a 5 year-old the
half- time is taken to be 55 days [89I1].
In cases of accidental intakes, Prussian Blue (ferric ferrocyanide) can increase the rate of excretion of
137
Cs from the body (see Chapter 9). If Prussian Blue is ingested it will accumulate any caesium secreted
into the gut preventing it being re-absorbed. The half-time of retention can be reduced to about 40 days;
the rate of loss is dependent upon the turnover rate of 137Cs in tissues and its loss into the gastrointestinal
tract.
137

Ruthenium
106

Ru is also produced in nuclear fission. Its absorption from the gastrointestinal tract is quite low; the
value for gut absorption (f1) is taken to be 0.05. The distribution of 106Ru in mice, rats, monkeys and dogs
is fairly uniform throughout all tissues after an initial period during which the kidneys contain the highest
concentration. The animal data have been used by ICRP [89I1] to define a retention function for
Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-17

ruthenium in man. Any ruthenium entering the blood is assumed to be distributed throughout the body
with retention components of 35 % (T = 8 days); 30 % (T=35 days) and 20 % (T=1000 days). The
remaining activity entering the blood (15 %) is taken to be promptly excreted.
Because of the relatively short retention time in the body of most of the intake and a physical half-life
of 368 days, the effective dose from ingestion of 106Ru is dominated by the radiation dose to the large
intestine from beta-particles emitted while it passes through the GI tract.
7.2.5.2 Elements that deposit mainly in particular organs or tissues
Iodine
Radioactive isotopes of iodine are important components of nuclear fission, particularly in the first few
days and weeks after a release into the atmosphere. If taken into the body they are accumulated by the
thyroid gland, as was demonstrated particularly after the accident at Chernobyl. The most important
isotope is 131I which has a physical half-life of 8.04 days. Radioactive isotopes of iodine are also widely
used in medicine. They are used to demonstrate changes in thyroid function, to treat hyperthyroidism or
to kill tumour cells in the treatment of thyroid cancer.
The thyroid gland consists of a bi-lobed body in the neck region. It produces the hormones thyroxine
and tri-iodothyronine which are important for regulating the body's metabolic rate. Disorders of the gland
can result in either an under- or over-active gland (hypo- or hyperthyroidism). The gland weighs about
20 g in the healthy adult (2 g in the newborn child) and is made up of 20 to 40 million spherical vesicles
(follicles) per lobe. Each follicle is surrounded by a single layer of cuboidal epithelial cells (acinar cells)
lying upon a basement membrane and in proximity to numerous blood capillaries. The vesicles are filled
with a structureless semi-fluid material the so-called colloid which contains the active component of
the gland (a protein-storage form of hormone). When the thyroid is quiescent, colloid is abundant and the
follicles large (about 300 m in diameter). When the thyroid is active colloid is scanty and follicles small
(about 50 m in diameter). The gland contains about 10,000 g of iodide in the average normal adult.
For adults in Europe about 225 g of stable iodide enters the extracellular (iodine) space from the diet
each day, absorption occurring across the small intestine within 1-2 hours. About 70 g of iodide per day
is trapped by the thyroid and converted to thyroid hormones while most of the rest is excreted in the
urine. The amount of ingested iodide that is taken up by the gland is thus about 30 % [83S1, 87S1]. The
fractional absorption varies between different individuals and there are significant differences between
countries because of varying levels of stable iodine in the diet. For the purpose of dosimetric modelling
ICRP has recommended that the uptake of radioiodine by the gland should be taken to be 30 %. The
iodide synthesised into hormone leaves the gland with a half-time of about 80 days (adult) and enters
other tissues. From this source most of the iodine (about 80 %) is metabolised back to free iodide with a
half-time of about 8 days and re-enters the iodide space, the rest is excreted in the faeces. In adults the
total amount of stable iodine excreted is approximately equal to the amount absorbed.
Iodide or elemental radioactive iodine (e.g. 131I) may be ingested (f1 = 1.0) or the volatile compounds
inhaled. As the gland is small, and it takes up about 30 % of radioiodine entering the blood, the
concentration of radioiodine in the gland, and hence the radiation dose, is more than a thousand times that
to other tissues. The turnover of stable iodine, and hence radioiodine, is low (T = 80 days in adults) and
thus short-lived isotopes (e.g. 131I T = 8.0 days) will decay mainly in the gland rather than being
returned to the blood. In children, although the turnover rate is faster the mass of the gland is smaller and
hence for a similar intake of radioiodine the dose can be higher. Milk consumption is the most important
pathway for the uptake of radioiodine from the human food chain after a release of radioiodine into the
environment and children have a high consumption of milk. As a consequence children are the most
sensitive (critical) group following such a release.
Various drugs have been used to reduce the uptake of radioiodine into the thyroid gland after an
intake. The safest and most effective procedure is to administer a large single oral dose (20-200 mg) of
potassium iodide or iodide (see Chapter 9). It is effective within an hour and reduces the subsequent
uptake of radioiodine into the gland. The daily intake is suddenly increased from about 225 g to
Landolt-Brnstein
New Series VIII/4

7-18

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

20-200 mg. However, the uptake of iodide into the thyroid remains at about 70 g d1 so that only a small
fraction of the stable iodine and hence radioiodine present in the iodide space is then transferred to the
gland. Thus, if 20 mg of stable iodide is given promptly the fractional uptake of radioactive iodine by the
gland can be reduced to less than 0.001 (0.1 %). Since the half-time of iodide in the iodide space is about
10 hours, with rapid uptake by the gland or excretion from the body, the effectiveness of administering
large amounts of stable iodide diminishes the greater the delay; after 48 hours, it is of little value at all. It
has no effect on any radioiodine that has been taken up by the gland. Administration of stable iodide is
the treatment of choice for accidental intakes of radioiodine and is recommended by the World Health
Organisation [99W1].
Alkaline earth elements
The bone is a highly specialised form of connective tissue. It consists of a soft organic matrix of collagen
and ground substance in which is deposited calcium hydroxyapatite (Ca10 (PO4)6 (OH)2). The cells of
bone consist of a proliferating population of stem cells which differentiate into: osteocytes the cells
responsible for bone maintenance; osteoblasts the cells responsible for bone formation; and osteoclasts
the cells responsible for bone removal. There are two types of bone:
Hard cortical bone, which makes up about 80 % of the bone mass and which is penetrated by blood
vessels and Haversian systems; and
Trabecular bone or spongy bone which makes up the remaining 20 % of the bone mass. The
trabeculae are generally 100-200 m in diameter and usually do not contain blood vessels. The spaces
between the trabeculae are large (up to 1000-2000 m) and contain the vascular (red) marrow. The
surface area of trabecular bone is estimated to be about 4 times that of cortical bone [79I1].
The surfaces of bone are covered with non-mineralised layers of connective tissue. This is the
periosteum on the external bone surface and the endosteum on the internal surface. The processes of bone
turnover, resulting from the laying down of new bone and removal of old bone, continue throughout life
although slowing down with increasing age.
Calcium is thus an important component of the skeleton and other alkaline earth elements can be
substituted for it in the bone matrix. A number of substitutions are possible in the lattice structure without
disturbing the symmetry of the crystal lattice. ICRP has developed age-dependent biokinetic models for
the alkaline earth elements (Ca, Ba, Sr and Ra) [93I1]. These models allow for the recycling of
radionuclides between the skeleton, blood and soft tissues. They are physiologically based and take
account of bone growth and remodelling as a function of age. They can also be applied to the
interpretation of bioassay data. These models are essentially dynamic in nature. Material initially
deposited on bone surfaces may be buried in bone volume by the formation of new bone or resorbed to
red bone marrow. Activity in marrow is either locally or systemically recycled back to bone surfaces, and
is also replenished by resorption from bone volume. A fraction will also be excreted from the body. The
rate at which some of the processes operate can depend on the type of bone, as well as age, and allowance
is made for this by division into cortical and trabecular compartments. This division also accommodates
age-dependent dosimetry in adults most of the active marrow is associated with trabecular bone,
whereas for children this is more evenly distributed between both types of bone.
7.2.5.3 Elements that deposit in a number of tissues
Plutonium, americium and curium
Plutonium and other higher actinides are produced in thermal reactors, although the relative amounts
generated depend upon the irradiation time. At low irradiation times almost pure 239Pu\240Pu are produced
(subsequently given as 239Pu). As the irradiation time increases other isotopes of plutonium (e.g. 241Pu)
Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-19

and higher actinides (e.g. 241Am, 242Cm, 244Cm) are produced. 239Pu is the isotope that has been processed
in greatest quantity and for which most biological data are available. 238Pu is also an important isotope
and is used as a power source in satellites and in cardiac pacemakers. Plutonium metal is highly reactive,
oxidising in moist air if present in a finely divided form. The oxide is chemically inert and insoluble,
particularly if produced at a high temperature (1000 C). Am and Cm metals readily react with oxygen to
produce oxides which are much more soluble than PuO2. In the processing of nuclear fuel soluble nitrates
and other soluble complexes are formed.
The behaviour of plutonium in the body depends upon its chemical form. Plutonium dioxide is
chemically inert and largely insoluble in biological fluids, particularly if produced at a high temperature.
Soluble plutonium compounds (e.g. plutonium nitrate, plutonium citrate) readily hydrolyse at the pH of
biological fluids. Following hydrolysis, there is a strong tendency to polymerise, forming a colloidal
insoluble compound. Alternatively, soluble compounds can react with naturally occurring complexing
agents in body fluids which can readily move around the body. Which of these reactions predominates
depends upon the site of entry of plutonium. If into blood, then complex formation is likely; if into a
wound or the lung, then colloidal polymers are likely to be formed.
The main route of entry of plutonium into the body is by inhalation, although it can also enter through
cuts and wounds. In adults very little is absorbed from the gut (ICRP has adopted gut absorption,
f1, values of 5 104 for soluble plutonium compounds and plutonium in foodstuffs, 1 104 for
plutonium nitrate, and 1 105 for plutonium oxide, 239PuO2) or across the intact skin. For americium and
curium ICRP has adopted an f1 of 5 104 for all compounds [94I1, 96I1].
After inhalation, deposition of plutonium in the lungs is determined by particle size as detailed in
Section 7.2.1.1. Subsequent clearance from the lungs depends upon its chemical form. Whatever the
chemical form inhaled, a fraction, consisting of any soluble material will be rapidly transported to blood
and this is excreted through the kidneys or deposited in tissues (mainly the liver and skeleton). The
remaining material, consisting of colloidal polymers or material with a low solubility (e.g. PuO2), is
initially retained in the lungs. Material retained in the lungs is largely taken up by scavenger cells
(macrophages) in the lungs. These cells may migrate to lymph nodes or reach the muco-ciliary escalator,
be swallowed and excreted in the faeces. Alternatively materials in macrophages may gradually dissolve
and translocate to blood. In studies with experimental animals retention half-times of the long-term
component of retention have varied from 100 to 1000 days or more although soluble compounds are
cleared more rapidly. The relative proportions of the long and short retention components depend upon
the material initially deposited. For example, in the case of a polydisperse aerosol of high temperature
calcined plutonium dioxide deposited in the lungs the amount rapidly moving to blood is normally less
than 0.4 % and would be treated as default absorption Type S in the HRTM (Section 7.2.1.2). For a
plutonium nitrate aerosol it may be greater than 20 % and would be treated as Type M. Compounds of
americium and curium, particularly the oxides, are more soluble than plutonium compounds in the
respiratory system and more readily absorbed. They would generally be classified as Type M but any
americium or curium trapped in insoluble particles of PuO2 would behave as Type S.
The principal sites of deposition of Pu in the body after translocation to the blood are the liver and
skeleton. This is also the case for Am and Cm. All three actinides are classified as bone-surface seeking
elements. That is, they are assumed to be uniformly distributed on endosteal bone surfaces of cortical and
trabecular bone after their deposition in the skeleton. In practice, surface deposits of Pu, Am and Cm have
been shown to be progressively buried by the formation of new bone. In addition, activity is lost from
bone surfaces during resorption and some transfer to bone marrow takes place, particularly for Pu. In
ICRP Publication 67, a dynamic model has been adopted to describe their behaviour in the body and to
take account of their movement in bone as well as between tissues and excretion (Fig. 7.7 [93I1]).

Landolt-Brnstein
New Series VIII/4

7-20

7 Internal dosimetry of radionuclides

Other
soft
tissue

Intermediate
turnover
(ST 1)

Slow
turnover
(ST 2)

Rapid
turnover
(ST 0)

Skeleton

Liver
Cortical
surface

Cortical
marrow

Trabecular
surface

Trabecular
marrow

Cortical
volume

Liver 2
Liver 1

Blood
Trabecular
volume

[Ref. p. 7-68

GI tract
contents
Kidneys
Urine

Urinary
bladder
contents

Other kidney
tissue

Gonads

Faeces

Urinary path

Fig. 7.7. Diagram of the biokinetic model for plutonium and


americium; [93I1].

Comparisons of relative skeletal retention in the biokinetic model are complicated by the compound
structure of the skeleton and recycling between compartments which gives rise to several retention
components. However, calculation shows that after short-term losses are complete it is possible to discern
an effective retention half-time in the skeleton of nearly 100 years (Fig. 7.8). The figure also gives the
retention of plutonium in the skeleton for 3-month-old infants and 10-year-old children. Because of the
faster turnover of bone at younger ages, the initial uptake of plutonium by the skeleton is greater although
the rate of loss is also faster. Fig. 7.9 gives comparable data for the liver. Although there are differences
in the initial uptake, reflecting the effect of age and deposition in the skeleton, overall liver retention is
similar for all age groups. The peak in uptake at 5-10 years reflects the uptake of activity lost to the blood
from the skeleton.
60

60

Infant
Age 10y
Adult

50

Infant
Age 10y
Adult

Injected Pu in liver [%]

Injected Pu in skeleton [%]

80

40

40
30
20

20

10

10

30
20
Years after injection

40

50

Fig. 7.8. Model predictions of the plutonium content of


the skeleton as a function of age and time after injection.

10

30
20
Years after injection

40

50

Fig. 7.9. Model predictions of the plutonium content of


the liver as a function of age and time after injection.

Intravenous injection of the complexing agent diethylenetriaminapentaacetic acid (DTPA) as the


calcium zinc salt is the only accepted therapeutic method for removing soluble forms of actinides from
the body. It forms chelate complexes with these actinides which can be excreted in the urine and hence
Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-21

can effectively clear them from the systemic circulation and some that has recently deposited in bone and
other tissues. It is unable to remove intracellular deposits or activity that has been buried in bone and
must, therefore, be administered soon after an intake. It can remove some soluble complexes from the
lungs. Local injection of DPTA into contaminated wounds can remove more soluble forms of plutonium
from the body than the same amount given intravenously, provided the DPTA completely infiltrates the
wound site. Further information on DTPA treatment is given in Chapter 9.

7.2.6 Excretion
In the biokinetic model described in ICRP Publication 30, no specific information was given on excretion
in urine and faeces, although the models were used in Publication 54 [88I1] for interpreting excretion
data. In the 1990 recommendations of the ICRP [91I1], however, the urinary bladder and the colon are
given explicit tissue weighting factors wT (see Section 7.3.1) and the revised biokinetic models given by
ICRP now give specific information on excretion pathways in urine and faeces [93I1]. For assessing
doses from systemic activity lost into the faeces, the model for the gastrointestinal tract is used (Section
7.2.2) assuming secretion of radionuclides from the blood into the upper large intestine. A model for the
urinary bladder has been adapted for calculating doses to the bladder wall [93I1]. The bladder is taken to
be of fixed size containing 15, 25, 65, 75, 85 and 115 ml of urine in the 3-month-old, 1-, 5-, 10-, 15-yearold children and adults, respectively. These volumes represent the average content of the bladder during
the time period between voids. The rate at which radionuclides enter the bladder is based on their
elimination rates from body tissues and the urine to faecal excretion ratio adopted for the biokinetic data
for each element. For some elements, the biokinetic data directly address excretion. The number of voids
per day for the 3-month-old and 1-year-old are taken as 20 and 16, respectively. For all other ages, 6
voids per day are assumed.

7.2.7 Embryo and foetus


During pregnancy, radionuclides that have entered the mother's body, either before or after conception,
can irradiate the developing embryo and foetus. The radiation dose to the offspring will depend upon a
number of factors. These include:
the transfer of radionuclide(s) to the developing offspring from maternal blood and from deposits in
the tissues of the mother;
the distribution and retention in foetal tissues;
the physical half-life and formation of decay products;
growth of the offspring; and
photon irradiation from radionuclides in the placenta and maternal tissues.
Radiation doses will also be received by the newborn child from radionuclides retained at birth.
During the foetal period of development, radionuclides can cross the placenta to reach the tissues of
the embryo and foetus from the maternal circulation. The processes involved in this transfer may include
simple diffusion, facilitated transport, active transport, movement through pores and channels, and
pinocytosis [83S1, 87S1]. Most human data on the placental transfer of radionuclides are available from
studies with labelled metabolites, radiopharmaceuticals and other radionuclides used in nuclear medicine,
although some data are also available for radionuclides in fallout from weapons testing or for radionuclide
releases into the environment as a result of nuclear accidents (e.g. 90Sr, 131I, 137Cs). Analysis of autopsy
samples has also given information on both naturally occurring and artificially produced radionuclides.
Information is additionally available on levels of stable elements in the placenta and foetal tissues that can
be compared with those in the adult. The rather limited amount of human data available has made it
Landolt-Brnstein
New Series VIII/4

7-22

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

essential to use the results of animal studies in the development of dosimetric models for man, although
even here information can be very limited for many elements. Chemical analogy is also of value in model
development.
ICRP has issued Publication 88 [01I1] giving dose coefficients for the embryo and foetus following
intakes of radionuclides by the mother. It covers selected radionuclides of the 31 elements covered in
Publications 56, 67, 69 and 71 (Table 7.1) and applies to the offspring of both members of the general
public and workers. In the development of biokinetic and dosimetric models, two approaches have been
used. Where sufficient information is available, element-specific models have been given. This applies,
for example, to tritiated water, caesium, iodine and the alkaline earths. When appropriate human data are
not available, animal studies have provided the main basis for model development using a generic
modelling approach.
It has been assumed, in the absence of more specific information, that the dose to all tissues of the
embryo, taken to be up to the end of 56 days after conception, (i.e. the end of the second month of
gestation), can be approximated by the dose to the maternal uterus. All organs and tissues of the
developing embryo thus receive the same dose.
The general approach that has been adopted for calculating equivalent doses to the organs and tissues
of the developing foetus from experimental studies in animals is to use average concentrations of a
radionuclide in the foetus (CF) and mother (CM) obtained shortly after injection. Where possible the value
of the CF:CM concentration ratio adopted has been based on results obtained in a number of different
animal species. The total activity transferred to and retained in the foetus from 57 days of gestation to
birth at 38 weeks (266 days) is calculated for each radionuclide from the CF:CM ratio. This ratio at the
time of the intake is assumed to stay constant for the remaining period of the pregnancy. This is taken to
be a conservative assumption. Some examples of (CF:CM) concentration ratios adopted are given in
Table 7.7. The concentration ratio may depend upon the time of the intake in relation to the start of the
pregnancy. Thus for an acute intake of plutonium at any time before pregnancy the CF:CM ratio is taken to
be 0.03; this ratio is then maintained at this value through the period of gestation. For an acute intake
during the first trimester of pregnancy, however, a ratio of 0.1 is adopted, subsequently increasing to 0.3
for an intake at the end of the second trimester (180 days) and 1.0 for an intake at term (266 days). Again
this ratio is assumed to be kept constant over the remaining period of the pregnancy.
Table 7.7 Concentration ratios for elements in the foetus and mother (CF:CM) following intakes by the
mother before or during pregnancy and corresponding ratios for the placenta (CPl:CM)

Element
H in HTO
Organic carbon
Sulphur
Zinc
Zirconium
Ruthenium
Caesiuma
Cerium
Plutonium
Americium

CF:CM
Intakes prior to
pregnancy
1.6
1.5
1
2
0.2
0.01
1
0.01
0.03
0.01

Intakes during
pregnancy
1.6
1.5
2
2
0.2
0.2
1
0.05
0.1/0.3/1b
0.1

CPl:CM
1
1.5
2
1
1
0.1/2c
1
0.1/1c
0.1/5c
0.1/2c

a Half-time of long-term component in mother during pregnancy taken to be 50 days


b Intakes in 1st and at the end of the 2nd/3rd trimester (see text)
c Intakes before/during pregnancy
Dosimetric models were developed by ICRP that allowed for the calculation of doses to the embryo
and to foetal tissues from radionuclides deposited either in the tissues of the embryo/foetus, in the
placenta or in the mother. To provide data that could be used for assessing a range of possible intake
scenarios dose coefficients have been given for acute and chronic intakes by the mother at various times
Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-23

both before conception or during pregnancy. Dose coefficients for the offspring following ingestion of
radionuclides by the mother are given for a range of f1 values in Publication 88, while dose coefficients
for the offspring after inhalation of radionuclides by the mother are given for both 1 and 5 m AMAD
aerosols (the default particle sizes for members of the public and workers, respectively) and appropriate
lung absorption Types.
For acute exposures, intakes of radionuclides are taken to occur at the time of conception and after 5,
10, 15, 25 and 35 weeks of the pregnancy and at 6 months and 2 years before conception. For chronic
exposures, intakes are taken to occur during the year of pregnancy, starting from conception and for
1 year or for 5 years prior to conception. This range of intake scenarios was selected to allow doses to the
offspring to be calculated for any pattern of intake by the mother. Equivalent doses to the date of birth
have been given in Publication 88 for the brain, for the most sensitive 8-15 weeks of gestation, and for the
tissue receiving the highest dose. The effective dose to birth has also been given using the wT values
recommended by ICRP in Publication 60 [91I1]. Whilst these values are not strictly appropriate for
exposures in utero, they have been used as no alternative weighting factors are available and the
calculation of effective dose provides a useful quantity for comparison with doses to the reference adult.
Effective doses (to age 70 years) received after birth are also given, together with the total committed
effective dose (before and after birth) received by the offspring.
The total committed effective dose to the offspring, eoffspring due to maternal intake of the radionuclide
is the sum of the effective dose received during the in utero period, ein utero and the committed effective
dose during the subsequent 70 years of post-natal life, epostnatal. That is:
eoffspring = ein utero + epostnatal

(1)

where
ein utero =

8w
0

h&uterus (t )dt +

w
T

38 w
8w

h&T (t )dt

(2)

and
e postnatal =

w
T

70 y
birth

h&T (t )dt

(3)

where the limits of integration in the first two integrals are in weeks and in the last term is in years. The
value h&T is the equivalent dose rate to individual tissues of the offspring during foetal life and after birth.
In the case of the embryo the dose to the tissues of the uterus is taken as a surrogate for the dose to the
embryo.
In conjunction with the preparation of ICRP Publication 88 a CD-ROM has been issued which gives
much more detailed information than in the publication [02I2]. In addition to the doses given in
Publication 88 it provides equivalent doses to 15 organs and tissues in the foetus as well as an average
equivalent dose to the remainder tissues. It also gives doses to the offspring to a number of times after
birth (10, 20, 40, 70 years). Additionally, doses have been given for a range of ten inhaled particle sizes.
Publication 88 gives only dose coefficients to the offspring and no information is provided on
comparative doses to the adult. Such a comparison has, however, been published [02S1]. The main
findings are that, in general, doses to the offspring are similar to or less than those to the reference adult.
For a few radionuclides the dose to the offspring can exceed that to the adult. This applies to 3H, 14C, 35S
and 59Fe, to radioisotopes of I and to radioisotopes of the alkaline earth elements including 45Ca, 89Sr,
90
Sr, 224Ra and 226Ra. For radioisotopes of iodine and the alkaline earth elements, the greatest doses result
from intakes during the last trimester of the pregnancy when there is the greatest foetal demand for iodine
and calcium. Whilst in most cases the doses to the offspring for the radionuclides covered in Publication
88 exceed those to the reference adult by a factor of about 2 to 3, in the case of some bone-seeking
radionuclides the difference can be around a factor of 10 for intakes of short lived isotopes towards the
end of the period of pregnancy. Some illustrative dose coefficients for the offspring following inhalation
of 137Cs by the mother (as a member of the public) are given in Table 7.8. In this case the doses to the
Landolt-Brnstein
New Series VIII/4

7-24

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

offspring are highest for intakes early in the pregnancy as they reflect doses received by the mother over
the period of the pregnancy. For intakes later in the pregnancy doses are lower as, although some 137Cs
will be retained in the newborn child, the half-time of retention is less than in the mother.
Table 7.8 Dose coefficients [Sv Bq1] for the offspring from acute intakes by inhalation of 137Cs
(T = 30 y) by the mother, as a member of the public (AMAD = 1 m, absorption Type F, f1 = 1.0).
Reference adult = 4.6 109 Sv Bq-1
Scenario
[weeks]a
130b
26
0c
5
10
15
25
35
a
b
c
d
na

Highest organ
dose (in utero)d
7.0 1013
6.2 1010
2.5 109
2.4 109
2.3 109
2.2 109
1.7 109
6.1 1010

Brain
(8-15 weeks)
1.9 1013
1.7 1010
6.7 1010
1.1 109
9.4 1010
na
na
na

ein utero
7.0 1013
6.2 1010
2.5 109
2.4 109
2.3 109
2.2 109
1.7 109
6.1 1010

epost natal
1.1 1014
9.6 1012
1.5 1011
2.5 1011
4.1 1011
6.7 1011
1.8 1010
4.7 1010

eoffspring
7.1 1013
6.3 1010
2.5 109
2.4 109
2.3 109
2.3 109
1.9 109
1.1 109

Intake at indicated time; negative times are prior to pregnancy


130 weeks = acute intake 2.5 years before conception
0 = acute intake at time of conception
For 137Cs all tissues receive the same dose
Not applicable

7.2.8 Transfer in maternal milk


Models are presently being developed by ICRP for the transfer of radionuclides to mothers milk that will
allow the calculation of dose coefficients for intakes by the offspring [03H1]. The publication will cover a
review of biokinetic data relevant to an assessment of the transfer of radionuclides to breast milk
following intake by the mother, the development of models, and the calculation of doses to the newborn
child resulting from the transfer of radionuclides to milk after inhalation or ingestion by the mother. It is
assumed that lactation lasts for a period up to 6 months after birth and that milk consumption increases to
800 ml d1 over the first week and then remains constant to the end of lactation. Doses to the infant will
be given for a range of intake scenarios. It is proposed that in the publication dose coefficients will be
given for acute intakes by the mother at 26 weeks before conception, for intakes during pregnancy at 5,
15, and 35 weeks after conception and for intakes after birth at 1, 10 and 20 weeks of age. In addition
doses from protracted exposures throughout pregnancy and lactation will also be included. These dose
coefficients should give a sufficient amount of information to understand the implications for doses to the
offspring for intakes at various times either before or after birth. Data for additional acute intake times
and for chronic exposures as well as for inhalation of a range of particle sizes will be included on a
CD-ROM. The dose coefficients for intakes by the 3-month-old infant given in previous publications
[96I1] will be used to calculate the doses from the intakes by the suckling infant in milk.
Some preliminary information is given in Table 7.9 (from 03H1) which gives the ratios of infant
(offspring) dose to adult dose for chronic intake of various radionuclides throughout pregnancy and
lactation. The values for lactation include contributions from activity retained in the mother from intakes
during pregnancy as well as transfer to milk from intakes by her during lactation. The results of
preliminary model calculations showed that intakes during pregnancy contribute an estimated 15 % of
activity in milk for 137Cs, 210Po and 241Am, about 10 % for 45Ca and 90Sr, 4 % for 239Pu and negligible
amounts for 131I. Doses to the infant from milk consumption are estimated to exceed adult doses in the
cases of 45Ca and 131I. Very similar ratios of infant to adult dose are obtained when considering acute
maternal intake by ingestion, during early lactation; that is, for maximum transfer to milk.
Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-25

Also shown in Table 7.9 are ratios of dose to the offspring and adult from activity transferred to the
embryo and foetus during pregnancy. These ratios are based on the dose coefficients for the offspring
given in ICRP Publication 88 [01I1]. These offspring doses from in utero exposure include contributions
from activity retained in the tissues of the child at birth, ranging from about 90 % of the total foetus
dose for 239Pu to less than 10 % for 137Cs. Only in the cases of 131I and 210Po do the doses to the offspring
from transfer in milk exceed that resulting from in utero transfer.
Table 7.9 Comparison of doses following chronic maternal ingestion of radionuclides throughout
pregnancy and lactation
Ratio of offspring : adult dosea
Radionuclide
Foetusb
Infant in milkc
45
Ca
12
2.7
90
Sr
1.5
0.8
131
I
1.0
2.4
137
Cs
0.4
0.4
210
Po
0.1
0.2
239
Pu
0.04
<103
241
Am
0.01
<103
a Committed effective dose (environmental exposures).
b Includes doses received in utero and from activity
retained by the child at birth (based on 01I1)
c Includes doses from activity transferred to milk as a result
of maternal intakes during pregnancy and lactation (preliminary calculations).

7.3 Dosimetric models


7.3.1 Introduction
The dose to organs of the body (this set of organs is referred to as target organs or target regions) depends
in part on the distribution of the activity within the body (this set of anatomical regions is referred to as
source regions) and the transport of the radiations emitted in nuclear transformations (decays) of the
radionuclide residing in the source regions. In general, the target regions as well as source regions
will be identified as organs of the body, but this need not be the case when knowledge suggests otherwise.
For example, the short-lived decay products of radon, when inhaled, deposit on the surfaces of the
respiratory airways from which they irradiate the basal cells of the bronchial epithelium as a target region
of interest. Various procedures have been employed in computing the dose to target regions, given the
information on the distribution of activity within the body. In the late 1960s - early 1970s, however,
efforts were devoted to establishing a unified formulation that would be applicable to all types of
radiations. The Medical Internal Radiation Dose (MIRD) Committee of the Society of Nuclear Medicine
led efforts to formulate a computational scheme based on physical framework in support of the dosimetry
of diagnostic radiopharmaceuticals. The formalism set forth by the MIRD Committee [76L1] deals with
the various radiations emitted in nuclear transformation of radionuclides in a consistent manner. This
formalism only addressed the absorbed dose quantity and was limited to short-lived radionuclides that
emit electron (beta and conversion electrons) and gamma radiations. However, the rigorous physical basis
of the system enabled it to be extended to the needs of radiological protection and the calculation of
equivalent dose by the ICRP in its Publication 30 issued in 1979.

Landolt-Brnstein
New Series VIII/4

7-26

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

In Chapter 4 the various radiological quantities are defined. The absorbed dose quantity D is a point
dosimetric quantity that can be defined in any material. The absorbed dose within the target region T of
the body is generally computed as the mean absorbed dose, denoted by DT , and thus is the average energy
per unit mass absorbed by the target region. The mean absorbed dose can, of course, be written as the
integral of point quantity D over the volume of the target. In radiological protection it is necessary to
address all the emitted radiations and to recognize that some radiations, and thus their contribution to
dose, are more biologically efficient than other radiations. Thus the basic dosimetric quantity of radiation
protection is the equivalent dose in target organ T and is denoted by HT. The equivalent dose is defined as
HT =

w D
R

T ,R

(4)

where the summation extends over all radiations R contributing to the mean absorbed dose DT ,R in the
target T. The radiation weighting factors wR represent judgments regarding the potential relative
biological damage of radiation R without regard to the specific tissue or health consequence. The values
in current use in dosimetry are given in Chapter 4, Section 4.5.2.2. The weighting factors reflect, in part,
the density of the ionization within the target which is indicated by the linear energy transfer (LET) of the
radiation. See Chapter 4 for further discussion.
The tissues of the body, of course, vary in their sensitivity to ionising radiation particularly with
respect to stochastic effects (cancer induction and hereditary disease). The effective dose quantity was
introduced into the radiological protection system in ICRP Publication 26 and was further amplified and
extended in Publication 60. The effective dose reflects the underlying information regarding the risk of
stochastic effects among the irradiated tissues and aggregates these contributions into a single dosimetric
quantity (see Chapter 4). The effective dose E is defined as
E=

w H
T

(5)

where the summation extends over the organs/tissues assigned tissue weighting factors wT as described in
Chapter 4, Section 4.5.2.4. Table 4.3 gives tissue weighting factors recommended in Publication 26
[77I1] and in Publication 60 [91I1]. The effective dose resulting from radionuclides within the body can
be added to that from external radiation fields to obtain a single quantity that encompasses both exposures
as described in Section 7.7.2.

7.3.2 Absorbed fraction and specific absorbed fraction


The formulation of a computational scheme that enabled the estimation of absorbed dose from all
radiations emitted in nuclear transformations of radionuclides was largely achieved by the introduction of
the absorbed fraction quantity. Consider a source region r within which radiation of type i is being
emitted. If target region v absorbs energy from the radiation emitted in the source region r, then the
absorbed fraction in v from r i (v r) is defined as the quotient of the energy imparted to the target
region v and the energy, exclusive of rest energy, emitted in the source region r. That is, the absorbed
fraction can be expressed as

i (v r) =

energy absorbed by target region v


energy emitted in source region r

(6)

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-27

The absorbed fraction is defined only for target regions which are volumes; however, no such
constraint is placed on the source regions, i.e., it could be a point, line, surface, or volume. The absorbed
fraction quantity embodies not only the geometric variables of size, shape, and spatial relationship of the
regions, but also the extent to which the radiation is transported through the medium containing these
regions.
The specific absorbed fraction i (v r) is defined as the absorbed fraction per unit mass of the
target region; i.e,

i (v r) =

i (v r)
mv

(7)

where mv is the mass of the target volume. The specific absorbed fraction has the property that it can be
defined for all target regions, including the important case of a point target. Recall from Section 7.3.1 that
absorbed dose is a point dosimetric quantity. There can, however, be no points in common between the
source and target region unless one of the regions is a volume.
If the source and target regions are in a homogeneous absorbing medium that is sufficiently large
(relative to the range of the radiation) that edge effects are negligible, and if the activity is uniformly
distributed in the source region, then the uniform isotropic model is said to apply. Under this model,
the distribution of absorbed energy about the source region is a function only of distance from the source.
The fraction of emitted energy absorbed per unit mass at a distance x can then be represented by the
point-isotropic specific absorbed fraction i ( x) . Since the emitted energy must be absorbed somewhere, the point-isotropic specific absorbed fraction must satisfy the constraint that

4 x 2 i ( x) dx = 1

(8)

where is the density of the homogeneous medium.


The point-isotropic specific absorbed fraction for the various radiations of interest provides the basic
means of estimating specific absorbed fractions. Non-point source and target regions can be developed
simply as the superposition of the point function. Thus the specific absorbed fraction between any nonpoint target region r and a point source P is the mean of the values of i (x ) in the target region

i (r P) = i ( x)

(9)

Furthermore, the specific absorbed fraction in any region r1 from a source in another region r2 is the
mean of the values of the point-isotropic specific absorbed fraction for all pairs of points in the regions;
i.e.,

i (r1 r2 ) = i ( x)

(10)

where x is the distance between points randomly selected in r1 and r2. The doubled-headed arrow in
equations 9 and 10 indicates that either region may be the source or target region. Equations 9 and 10 can
be expressed in their integral representation, but for regions whose geometry is complex, i.e., other than
spherical, recourse is often made to numerical evaluation using Monte Carlo techniques to randomly
select the points.
As noted above, the point-isotropic specific absorbed fraction is a function only of distance. Thus if
the source and target regions are interchanged in Equations 6 and 7, the numerical value of the specific
absorbed fraction does not change. This property of the uniform isotropic model is referred to as the
Reciprocity Theorem. The conclusion of the theorem is that the specific absorbed fraction i is
independent of which region is designated as the source and which the target is. In symbols
Landolt-Brnstein
New Series VIII/4

7-28

7 Internal dosimetry of radionuclides

i (r1 r2 ) = i (r2 r1 ) i (r1 r2 )

[Ref. p. 7-68
(11)

and therefore

i (v 2 v1 )
m2

i (v1 v 2 )
m1

(12)

Note that these relationships apply to all radiations in the uniform isotropic model.

7.3.3 Computational models of the human anatomy


7.3.3.1 Mathematical phantoms
The application and extension of the above in radiological protection necessitated the formulation of a
computational model of the human body a so called mathematical phantom. Such a mathematical
description of the adult human was used to provide estimates of the absorbed fractions for photon emitters
distributed in the model [69S1]. Much of the stimulus for this development came from the needs of the
MIRD Committee and thus the mathematical model is often referred to as the MIRD phantom although
over the years many modifications have been made to the model including the extension to children
(Fig. 7.10).
The MIRD phantom consists of three principle Sections: (1) an elliptical cylinder representing the
arms, torso, and hips,; (2) two truncated elliptical cones representing the legs and feet; and (3) an
elliptical cylinder representing the neck region and lower portion of the head, which is topped by a half
ellipsoid. The organs of the body were represented by simple conic sections in some cases cut by planes
and rotated. The defining equations were readily evaluated and thus the model was well suited for use
with Monte Carlo techniques in the computation of photon absorbed fractions. Limitations in the
computational hardware of the early 1970s, not the available anatomical information, restricted the
realism of the modelling.
7.3.3.2 Voxel models
Voxel models are human models based on computed tomographic or magnetic resonance images obtained
from high resolution continuous scans of a single individual. The greyscale data of the medical images are
interpreted into tissues (i.e. organs), a process known as segmentation. Each volume element, called
voxel, has an identification number that identifies the discrete organ of that particular voxel. The models,
consisting of millions of voxels, provide a three-dimensional representation of the human body and the
spatial form of its constituent organs and structures. They were initially developed for radiological
protection purposes to estimate the risk to a person or population due to an irradiation. For this purpose, a
detailed model of the human body is required, together with computer codes simulating the radiation
transport and energy deposition in the human body. They are at present the most precise representation of
the human anatomy to be used for computational radiation transport simulation.

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-29

Fig. 7.10. Schematic diagram of the MIRD phantom; [78S1]. Various modifications have
been made by other investigators; [87C1].

Among other laboratories, the GSF- National Research Centre in Germany started the development of
voxel models covering various ages and anatomical statures [02P1]: an 8 week old baby girl (Baby) and a
7 year old girl (Child) [88Z1], four male models (Golem (38 y), Visible Human (38 y), Frank (48 y) and
Otoko (40 y)) and three adult female voxel models named Donna (40 y), Helga (26 y) and Irene (32 y)
[02P1]. Fig. 7.11 shows three dimensional reconstructions of some organs of the above female models
and demonstrates their anatomic realism. The male adult phantom Golem [01Z1] has height and weight
similar to the ICRP Reference Man [75I1]. Otoko (01S1) stems from whole body CT data of a patient
whose external dimensions are in good agreement with the Japanese Reference Man [89T1]. The Visible
Human was constructed from CT data and photographic data from the Visible Human Project of the
American National Library of Medicine, obtained from the CT pictures of a donated body of an executed
38-year-old man from Texas, USA. Both Visible Human and Frank are rather broadly built, the former
being also very tall, and are probably more suitable to simulate bigger individuals. Similarly, Donna and
Helga are taller and heavier than the reference adult female, as characterised in ICRP Reference Man
[75I1] and ICRP Report 89 [02I3], whereas Irene has a weight below the reference female.
Due to their anatomical realism, such models have been the subject of increasing interest and
acceptance, and others have been developed elsewhere: Zubal et al [94Z1] at Yale developed a head and
torso phantom as well as a head phantom with fine resolution from the CT data of an adult male with
dimensions similar to the MIRD-5 mathematical phantom [78S1] who was imaged from head to midthigh. Dimbylow and his colleagues [96D1] at NRPB, UK, have developed the Norman model based on
whole-body MRI scan data of a healthy volunteer. The exact dimensions of the voxels were scaled so that
height and mass of the segmented model agreed with the values of the Reference Man [75I1]. A body
representation that gained recent popularity is the Visible Human Male [98S1]; this data set was
assembled using the original colour photographic anatomical slice images of the body mentioned above
Landolt-Brnstein
New Series VIII/4

7-30

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

which was acquired by the Visible Human Project of the American National Library of Medicine. Xu and
co-workers [00X1] at the Rensselear Polytechnic Institute have developed the VIP-Man based on these
data, which is the most complete body description so far with respect to number of structures defined and
voxel resolution. More recently, Kramer et al [03K1] created MAX based on the Yale model. Regarding
paediatric models, Caon et al [99C1] constructed a trunk model of a 14-year-old female and Nipper et al
[02N1] developed a newborn model which is based upon a high resolution CT scan of a 6-day female
baby.

Fig. 7.11. View of the skin and


some organs of the female models
Helga, Donna and Irene (left) and
of the skeleton and some organs
of the male model Golem (right);
[02Pet]. On the extreme right is
an image constructed from the
colour photographic anatomical
slice images of the Visible
Human Project of the American
National Library of Medicine;
[00X1].

Several image processing functions are used for segmentation of voxel models. However, none of
these methods works automatically; but interactive methods controlled by an operator must be applied
and thus the segmentation of a whole body model with several organs is a time-consuming procedure.
Moreover, it is difficult to obtain a suitable fine, contiguous data set.
The most important advantage of the voxel phantoms over mathematical phantoms, is their realism
concerning anatomy: the organ shape as well as the organ location is realistic, since computed
tomographic images from an actual subject were employed for their construction. Thus, they offer a clear
improvement compared to the older mathematical models whose organs are described by relatively
simple geometrical bodies. Furthermore, the distance between the organs, an important parameter,
particularly for internal dosimetry where several organs are the so-called source organs and all the others
are the targets, are realistically simulated by the voxel models, which is not always the case for the
MIRD-type models.
One of the most interesting characteristics of the voxel models is the possibility of varying their size
and hence simulating smaller or bigger individuals. Their external dimensions can be adapted for each of
the three dimensions independently. All internal dimensions of the resulting scaled-down or scaled-up
version of the original model are consequently modified with the same scaling factors. However, the
scaling factors should range within reasonable magnitudes, to avoid anatomical errors in the organ
proportions.
A limitation of the voxel models is that very small tissues, as for example the eye lenses and the skin,
cannot be represented with their correct thickness because it is not possible to segment structures below
the voxel resolution. An additional concern is that the supine position of the body during acquisition of
the image data results in an upward shift of the abdominal organs and the compression of the lungs.
However, for radionuclides with residence times in the body of a few days much of the dose is received
under various postures including sitting or lying down. The position of the stomach is well known to be
quite varied depending on its content.
Strictly speaking voxel models represent themselves and not a reference or an average individual.
For many situations in radiation protection, this is fully adequate, particularly if their external dimensions
comply more or less to the reference or the exposure conditions cannot be accurately described.
Moreover, since the models available range from very slim persons to large and heavy persons, they can
be used to estimate the doses to an individual by selecting the model fitting best to the person under
consideration. However, for certain situations involving regulations, guidelines or dose limitations, for
example exposures of a population or radiation workers, dose values are required for a reference
Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-31

individual. As a consequence, there is a need to construct body models that combine a realistic anatomy
with organ masses, shapes and locations that are representative. Probably the best way to achieve such an
aim is to modify an appropriate voxel model (i.e., one that already resembles Reference Man data in its
external body dimensions) to one having reference organ masses as well, retaining its realistic anatomy.
This approach was used by Dimbylow [96D1] to construct NORMAN, the model of an adult male, and is
now underway at the GSF-National Research Centre in Germany under the supervision of the ICRP.
These reference voxel models are then appropriate for calculating organ doses for reference persons and,
hence, establishing reference dose conversion coefficients for international recommendations such as
those from ICRP.

7.3.4 Dose rate per unit activity, S-factor


Consider an organ T of the body which at time t is absorbing energy from activity in a source region S of
the body. Let the activity, i.e., the average rate of nuclear transformations (nt), in the source region be
AS(t) and denote the mean energy emitted as radiation of type R per nuclear transformation by R; that is
R = YR ER, where YR and ER are the yield (number per nt) and energy of radiation R, respectively. The
rate at which energy is absorbed in T per unit mass at time t, which is by definition the mean absorbed
dose rate, D& (T S,t ) due to the activity in S is
R

D& R (T S,t ) = AS (t ) R R (T S)

(13)

In general, more than one type of radiation will be associated with the nuclear transformation
process of a particular radionuclide, and thus the mean absorbed dose rate is
D& (T S,t ) =

D&

(T S, t ) = AS (t )

R (T S)

(14)

If the activity is present in a number of source organs, then an additional summation must be
considered to derive the mean absorbed dose rate in T
D& (T ,t ) =

A (t )
S

R (T S)

(15)

where the first summation is over all source regions S.


Examination of the above equations reveals that the factors within the inner summation, i.e., R and
R(TS), reflect the physical data on the nuclear transformation process and the transport of the emitted
radiations between S and T which depend in part on nature of the radiations and the anatomical
relationship of these two regions. Given an agreed-upon analog of the human body for estimation of R,
then considerable effort can be saved through consideration of the additional quantity S. If we define
S (T S) as
S (T S) =

(T S) =

ER R (T S)

(16)

where YR is the yield of radiation R of energy E R . Then the expression for the mean absorbed dose rate
reduces to

Landolt-Brnstein
New Series VIII/4

7-32

7 Internal dosimetry of radionuclides

D& (T ,t ) =

A (t ) S (T S)

[Ref. p. 7-68
(17)

The quantity S represents the mean absorbed dose rate in T per unit radioactivity in S. If S is
considered to be invariant with time, that is, if the analog of the human body and its implied geometric
relationships are independent of age, then integration of Equation 17 yields the mean absorbed dose in T.
D (T ) =

S (T S)

(18)

~
where AS denotes the time integral of the activity in the source region (the cumulated activity). Thus S
may also be defined as the mean absorbed dose per unit cumulated activity. Methods for deriving the
cumulated activity are discussed in Section 7.4.1.
The S factor can be expressed in terms of equivalent dose by inclusion of the radiation weighting
factors in its defining equation; i.e,
SEE (T S ) =

wR R (T S) =

ER wR R (T S)

(19)

where wR is the radiation weighting factor (Section 7.3.1). The quantity SEE is a radiological
protection quantity introduced by ICRP (1979) as the specific effective energy. However a more
appropriate name would be the specific equivalent energy.
The equivalent dose rate to tissue T can be written in terms of SEE as follows,

H (T ,t ) =

A (t ) SEE (T S)
S

(20)

and the effective dose rate is defined as the sum of the weighted equivalent dose rates in a number of
tissues (Section 4.5.2.3 and Equation 5, this Chapter).

E (t ) =

H (T ,t )

(21)

7.3.5 Specific absorbed fractions for various radiations


The three principal modes of nuclear transformation are beta decay, alpha decay, and isomeric transition.
An additional process, spontaneous fission, is available to some heavy nuclides. The principle radiations
involved in these modes of nuclear transformation are alpha particles, electrons (either negative or
positive charge) and photons (electromagnetic radiation). The latter two radiations may arise from the
nucleus as well as the orbital electrons of the newly formed radionuclide. These radiations differ
significantly in their energy deposition pattern, as a result of different mechanisms through which they
interact with matter, further details are given in Section 3.5.

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-33

7.3.5.1 Electrons
A continuous energy spectrum of electrons is associated with beta decay. The spectrum ranges in energy
from zero to the maximum energy permitted by the difference in the energy level of the parent and
daughter nucleus. Electrons of discrete energy are also observed in nuclear transformation, as a result of
processes involving the orbital electrons.
Under the auspices of the MIRD Committee, Berger has tabulated point-isotropic specific absorbed
fraction data for monoenergetic electron sources ranging in energy from 0.025 to 5 MeV [71B1]. To
facilitate numerical use, Berger presented the data in terms of a scaled point kernel F(r/r0) where r0 is the
continuous slowing down approximation (csda) range. The point-isotropic specific absorbed fraction (r)
in terms of Berger's scaled point kernel is

(r ) =

1
F (r / r0 )
4r 2 r0

(22)

where is the density of the medium. The tabulations were prepared for water as a surrogate medium for
soft tissue. Table 7.10 presents the 90-percentile distance (x90) in water as a function of electron energy.
The 90-percentile distance is defined to be the radius of a sphere around a point source within which 90 %
of the emitted energy is absorbed. As can be seen from Table 7.10, electrons of energy up to about 2 MeV
deposit their energy within a distance of less than 1 cm.
Table 7.10. Deposition of electron energy (range-energy relationships). Percentile distance x90 in water
for electrons from monoenergetic sources. The results for E0 0.020 MeV are extrapolated (based on
Table 9, page 15, 71B1).
E0 [MeV] x90 [cm]
E0 [MeV]
x90 [cm]
4.0
1.57
0.70
0.207
3.5
1.36
0.60
0.169
3.0
1.16
0.50
0.131
2.6
0.99
0.40
0.096
2.2
0.82
0.30
0.063
2.0
0.74
0.20
0.0334
1.5
0.53
0.10
0.0106
1.2
0.41
0.05
0.00318
1.0
0.328
0.010
0.000194
0.90
0.287
0.005
0.000060
0.80
0.247
0.001
0.000008
Organs of the body are of dimensions sufficiently large relative to the electron range that the electron
absorbed fraction may be taken as unity if the source is uniformly distributed in the organ. Thus the
specific absorbed fraction for electrons is
1 / mT , if T = S

(T S) =

0 ,

if T S

(23)

A notable exception to the above occurs for walled organs where the source resides in the contents,
e.g., urinary bladder and the segments of the gastrointestinal tract.
For organs whose contents contain an electron emitter, the specific absorbed fraction in the wall of the
organ from its contents is usually taken to be given by

(wall contents) =
Landolt-Brnstein
New Series VIII/4

1
2 mcontents

(24)

7-34

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

where mcontents is the mass of the contents. This relationship is derived from the fact that the dose rate at
the surface of a half space containing a uniform distribution of activity is one-half the equilibrium dose
rate at locations within the contaminated half space far from the interface. It should be noted that the
approach for walled organs may be very conservative, in that the critical cells are typically considered to
be the basal cells of the epithelial layer, which lie at some depth in the tissue; in the gastrointestinal tract,
they are further shielded by a mucus layer. Thus the dose rate in the wall may decrease rapidly from the
value at the surface, particularly for low-energy electrons. Consideration of these details in the dosimetric
models must await further description of the location of the cells at risk.
7.3.5.2 Alpha particles
The point-isotropic specific absorbed fraction (x) has not been tabulated in the literature for alpha
particles since the range of alpha particles in tissue is sufficiently small that for organs of the body, an
absorbed fraction of unity can be assumed. However, in some specific instances such as alpha emitting
short-lived radon daughters deposited on the airways of the lung consideration must be given to the
energy deposition pattern.
The point-isotropic specific absorbed fraction (x) can be defined as

( x) =

1
(dE / dx) x
4 x 2 E

(25)

where (d E /d x) x is the stopping power of the alpha particle at the energy it has after travelling a distance
x, and E is the initial energy of the alpha emission. In order to avoid the discontinuity at x = 0, the
quantity 4x 2 ( x) is tabulated for the point-isotropic specific absorbed fraction. The mass stopping
1
(dE / dx ) and the csda range as a function of energy in soft tissue are shown in Figure 7.12.
power

























5DQJH JFP



G(G[   0H9FP J

These data can be used with Equation 25 to compute the point-isotropic specific absorbed fraction.
Specific absorbed fractions for source-target pairs in the body are the same as employed for beta
radiation, that is Equation 23 is applicable to solid organs. For walls of the gastrointestinal tract Equation
24 is applied, however only 1 % of the alpha particles' energy is considered to penetrate the mucous
lining of the tract.







Fig 7.12. Mass stopping power and range of


alpha particles in soft tissue.

( 0H9

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-35

7.3.5.3 Gamma-rays and characteristic X-rays


Gamma-rays and X-rays are electromagnetic radiations of short wavelength, orders of magnitude shorter
than visible light. A nucleus in an excited state from which it is energetically impossible to de-excite
through emission of particulate radiation (emission of alpha or beta particles) may de-excite through the
emission of one or more photons of electromagnetic radiation. Many nuclides formed in beta or alpha
decay may be in an excited state, and thus gamma-ray emission often accompanies these decays.
Electromagnetic radiations associated with changes in nuclear state are referred to as gamma radiation.
A measure of the probability per unit distance (density distance) travelled by a photon that an
interaction occurs is the mass attenuation coefficient. As the three interaction events, photoelectric effect,
Compton effect and pair formation (see Chapter 3) are independent and mutually exclusive, the total mass
attenuation coefficient / is given as

/ = / + / + /

(26)

where /, / and / are the mass attenuation coefficient for the photoelectric effect, Compton effect,
and pair formation interactions. Tabulations of these coefficients for various elements and compounds of
general interest have been given [95H1]. Values for other compounds or absorbing media can be
computed as

/ =

w ( / )
i

(27)

where ( /)i is the tabulated value for the ith element, and wi is the fraction by weight of the ith element
in the medium of interest. The equation is valid because the chemical binding energies between atoms in
a molecule are very small, thus not significantly altering the electronic binding energies. The transfer of
energy from the photon to secondary electrons is given by the mass energy-transfer coefficient and is
denoted by en /. The mass energy-transfer coefficient is the weighted sum of the mass attenuation
coefficients; i.e.,

en / = f ( / ) + f ( / ) + f ( / )

(28)

The weights f, f, and f indicate, for their respective interactions, the fraction of the photon energy
which is converted into kinetic energy of secondary electrons and dissipated in the medium by collision
losses. It is beyond the scope of this Chapter to detail the prescription for estimation of the weights. It is
important to note that the weights reflect only the energy transferred as kinetic energy of charged particles
and thus energy emitted as X-rays following photoelectric effect and the rest mass energy of the positronelectron pair in the pair formation process are excluded from the weight. It should be further noted that
for the composition of body tissues and typical photon energies, the correction for bremsstrahlung energy
loss by the secondary electrons is rather small.
7.3.5.4 Point-isotropic specific absorbed fraction
The fraction of the energy emitted by a point- isotropic source that is absorbed per unit mass at a distance
x from the source the point isotropic specific absorbed fraction (x) can be expressed as

( x) =

en e x
B ( r )
4 x 2 en

Landolt-Brnstein
New Series VIII/4

(29)

7-36

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

















 FP J







HQ FP J

where x is the distance from the point source, is the linear attenuation coefficient at the source energy,
en / is the mass energy-transfer coefficient at the source energy, and Ben(r) is the energy-absorption
buildup factor. The mass attenuation and mass energy-transfer coefficients for soft tissue as a function of
photon energy are shown in Fig. 7.13.
The build up factor is defined as the ratio of the absorbed dose obtained from a measurement to the
absorbed dose calculated to be due to uncollided photons at the location. The scattered photons are of
lower energy than the uncollided photons and hence subject to increased absorption as seen in Fig. 7.13.
Several tabulations of energy-absorption buildup data are available in the literature for application
to body tissues. Berger presented such data in MIRD Pamphlet No. 2 [68B1] for a point source in
water. Published values applicable to 40 mean free paths (r = 40) have been published by
Spencer and Simmons [73S1], whereas Berger's data were applicable to only 20 mean free paths.
For small values of r, Ben(r) is approximately unity and increases rapidly with increasing values
of r. The maximum value of the buildup factor occurs for photons of about 100 keV.



















Fig 7.13. Mass attenuation and mass energytransfer coefficients for photons in soft tissue;
[95H1].

( 0H9

7.3.6 Calculation of doses to soft tissues and the skeleton


For the majority of tissues in which radionuclides deposit, following their entry into the blood, the
assumption is made that both they and the sensitive cells are essentially uniformly distributed. On this
basis the average tissue dose is calculated. This applies for example to the liver, spleen, kidneys, muscle,
gonads and glands. In some cases, particles or colloids containing radionuclides may enter the blood and
these will result in a heterogeneous distribution of activity in tissues in which they deposit, particularly
for alpha emitters with a short path length (~ 50 m). Most experimental evidence suggests, however, that
a heterogeneous distribution of activity is no more likely to produce long term damage, such as cancer,
than a homogeneous distribution of activity and the calculation of average tissue dose is justified [80I3,
99S1, 03M1]. In the case of the skeleton, however, it has been necessary to take account of the way
radionuclides deposit in order to assess the dose to sensitive tissues.
A generalised model to assess doses from bone seeking radionuclides was given in ICRP Publication
30 [79I1] (Table 7.11). The target tissues in the skeleton are taken to be the active red bone marrow
(RBM), which is present in cavities in trabecular bone, and endosteal and epithelial cells assumed to lie
within 10 m of bone surfaces (BS). Energy deposited in the yellow marrow of cortical bone is not
considered to cause any radiation effects. The radiation dose to the RBM and BS depends upon the
pattern of deposition of the radionuclides in the bone (volume or surface deposition), the radiation it emits
(, , ) and the effective half-time. Some bone surface seeking elements are Ga, Zr, Th, Pu, Am and Cm
and some volume seekers Ca, Sr, Ra and U. The most recent biokinetic models for Pu, Am and Cm allow
for their progressive burial in the skeleton (Section 7.2.5.3).
Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-37

The total endosteal area of the skeleton over which the dose is calculated is taken as 12 m2, half
associated with cortical bone and half with trabecular bone. The layer on bone surfaces over which the
equivalent dose is averaged has a mass of 120 g. The mass of the red bone marrow in cavities within
trabecular bone is taken to be 1500 g. Table 7.11 gives the fraction of energy absorbed for - and emitters deposited in the skeleton. Thus, for an -emitter such as 239Pu, which initially deposits on the
bone surface, half the activity deposited in the skeleton is taken to be associated with trabecular bone
surfaces and half to cortical bone surfaces. For activity on trabecular bone surfaces 25 % of the energy
released will be absorbed by the sensitive 10 m cell layer on bone surfaces (BS), 50 % will be absorbed
in the red bone marrow (RBM); the remaining energy will be harmlessly dissipated in bone mineral. For
activity on cortical bone surfaces 25 % of the energy released will be absorbed by cells on bone surfaces;
the rest will be dissipated in yellow-marrow or in mineral bone. For radionuclides deposited in bone
volume the fraction of energy deposited in sensitive cells, and hence the dose, will be less than for bone
surface seekers as much of the energy will be dissipated in bone mineral. It should be noted that some
recent analyses of radiation-induced bone tumours and information on bone cell development suggests
that sensitive cells for bone tumour induction may reside at distances of more than 10 m from bone
surfaces and that a depth of 50 m may be a more appropriate depth over which to calculate doses
[00G1]. Preliminary calculations suggest that this may reduce the dose to cells near trabecular bone
surfaces by a factor of about 2.
Table 7.11. Fraction of energy deposited in target organs for - and
surfaces or in bone volume
Source organ
Target
-emitter
-emitter -emitter on
organ
uniform in
on bone
bone surfaces
volume
surfaces
E 0.2 MeV
Trabecular bone BSa
0.025
0.25
0.025
Cortical bone
BS
0.01
0.25
0.015
Trabecular bone RBMb
0.05
0.5
0.35
Cortical bone
RBM
0.0
0.0
0.0
a BS = Bone surfaces; b RBM = Bone marrow

-emitters deposited on bone


-emitter on
bone surfaces
E<0.2 MeV
0.25
0.25
0.5
0.0

-emitter
uniform in
bone
0.025
0.015
0.35
0.0

7.4 Dose coefficients


The previous Section described some of the fundamental principles used in the calculation of radiation
dose. In practice many radiation protection professionals will not need to implement these methods in
full, but will use published values of dose coefficients as a means of estimating doses to individuals or
populations.
A dose coefficient is defined as a committed dose per unit intake (Sv Bq1), the term can be applied to
either committed equivalent doses or committed effective doses. Dose coefficients are sometimes referred
to as dose per unit intake values. ICRP has adopted lowercase letters to denote dose coefficients. Thus,
a committed equivalent dose to a tissue T is denoted HT and the corresponding dose coefficient is denoted
hT, similarly the dose coefficients for effective doses (E) are denoted e. Dose coefficients provide a means
of converting intakes (Bq) into committed doses (Sv) from internal emitters and are therefore used in
many branches of radiological protection such as environmental assessments, the protection of workers
and nuclear medicine.
As referred to earlier in this Chapter, ICRP has, over the last decade or so, published a number of
documents giving dose coefficients for workers and members of the public, including children (Table
7.1). These Publications provide a consistent source of reference values. ICRP has also issued a
publication giving doses to the embryo and foetus from intakes by the mother. In addition ICRP has given
dose coefficients (usually expressed in mGy MBq1) for use in nuclear medicine. More details are given
in Section 7.4.2.4.

Landolt-Brnstein
New Series VIII/4

7-38

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

7.4.1 Method of calculation


The biokinetic models recommended by ICRP are described in Section 7.2. These models describe the
fate of an inhaled or ingested radionuclide as it enters the body, its subsequent absorption and distribution
among systemic tissues and its elimination from the body. Therefore, in most cases these models are
combined to form a complete model for predicting the behaviour of the radionuclide from initial entry
into the body until excretion or decay. For example, the complete model for inhaled plutonium would
require that the Human Respiratory Tract Model (HRTM) be combined with the systemic model for
plutonium (for activity taken up to blood) and the model for the gastrointestinal tract (for activity
mechanically cleared from the lung and swallowed and for activity excreted into the GI tract in bile).
These model constructs are referred to as compartment models, each compartment representing an
apparent influence on the kinetic behaviour of the radionuclide.
All the models recommended by ICRP to date describe the transfer of radionuclides (activity) between
compartments of the model by linear first order processes. That is, the rate of biological removal of
activity from a compartment at time t is taken to be the product of the activity in the compartment at time
t and a transfer rate coefficient, usually denoted by k. Consider an isolated compartment i as in Fig. 7.14.
Compartment i receives activity from and transfers activity to all other compartments in the model. The
element kj,i of the transfer rate coefficient matrix describes the fraction of the activity in compartment i
transferred to compartment j per unit time. The model is completely described by the transfer rate
coefficients matrix and the initial activity (content at time zero) in each compartment. The nuclear
transformations of many radionuclides form radioactive nuclei which must be considered in computing
the dose coefficient. Thus the intake of a radionuclide, the parent, may result in a series of radionuclides
being formed within the body. The kinetics of radioactive series must be superimposed on the kinetics
described by the biokinetic models. The activity Am
i of member m of the decay chain in compartment i is
given by a set of coupled linear differential equations of the form of Equation 30.
d m
Ai (t ) =
dt

inflow
n

j =1, j i

m
i, j

A (t ) + m
m
j

ingrowth
m1

m , m

m
i

outflow

A (t ) A (t ) (m +
m
i

m=1

(30)

m
j ,i

j =1, j i

where m = 1 for the parent radionuclide, k mj ,i is the fraction of the activity of chain member m in
compartment i transferred to compartment j per unit time, m is the decay constant of chain member m,
Fm, m is the fraction of the decays of chain member m which form member m (often referred to as the
branching fraction) and by definition Aim (t ) = 0 if m = 1 . In computing dose coefficients the set of
differential equations are generally solved as an initial value problem such as specified in Eq. 31.
0 for all i and m > 1

A (0 ) = 0 for m = 1 and i not a compartment of intake


nonzero for m = 1 and i as a compartment of intake

m
i

(31)

For example, an intake by ingestion would assume that at time zero one unit of activity of the parent
radionuclide is present in the stomach content and zero activity of the parent and daughter products is
present in all other compartments of the model.
Two approaches have been applied to describe the biokinetics of the daughter products. The so-called
shared kinetics assumes that the behaviour of the daughter is the same as the parent. That is,
kim, j = ki1, j , m = 2,..., n . If information indicates that the daughter product behaves differently from the
Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-39

parent radionuclide in the body then independent kinetics are applied in Equation 30. The well recognized
independent kinetic cases include the formation of noble gas radionuclides in the decay of solid daughter
83
parents, e.g., 35
Br8336m Kr and radioiodine formed in the decay of radioisotopes of tellurium, e.g,
Te133
53 I . However other cases of importance include the long decay series associated with some of the

133
52

natural decay chains (such as 22890Th ) where fundamentally different behaviours of the chain members in
the skeleton are encountered.
k i, j

Isolated compartment i
Ai (t)

k j,i

Fig 7.14. Isolated compartment exchanging activity with other


compartments in the model.

The biological removal coefficient of the activity of chain member m from ith compartment is given
n

by

m
j ,i

which can be stated as a half-time by dividing ln(2) by the removal coefficient. This would be

j =1, j i

the half-time for removal if there were no input into the compartment from other compartments. If the
compartment is subject to continued input then a plot of the compartment activity as a function of time
will reflect the removal rates from all compartments feeding the ith compartment. Thus in general one has
to be careful in speaking about biological half-times for complex models.
With the exception of a catenary model (a model in which the compartments only communicate with
adjacent compartments; that is, the nonzero members of transfer coefficient matrix are k1,2, k2,3, k3,4, ...,
kn1,n) no closed form solution exists for Equation 30. The catenary form of Equation 30 is well-known in
radiation protection as it describes the serial kinetics of a decay chain and its solution was formulated in
earlier work by Bateman in 1910 [10B1] who was assisting Lord Rutherford in the early investigations of
radioactivity. Catenary models were used in ICRP Publication 2 [59I1] and their solution has been
extensively investigated by Skrable [74S1].
Since only the catenary system or very small systems (less than four compartments) can be solved
exactly, it is necessary to approximate the solutions numerically. Applicable numerical approaches
include analysis of eigenvalues and discrete variable methods (also called step-by-step methods or
difference methods). With the advent of powerful desktop computing the discrete variable Runge-Kutta
method can be readily applied to Equation 30 (see for example 88B1). However well-written variableorder, variable-step routines such as the solver developed by Gear [71G1] are more efficient than a fixedorder, variable-step Runge-Kutta routine. The calculations of Publication 30 [80W1] were carried out
using Hindmarshs coding of the Gear method [74H1]. A collection of state-of-the-art solvers for the
initial value problem for ordinary differential equation systems, including the Gear method, are contained
in the widely available ODEPACK package [83H1]. Classical methods from linear algebra involve the
Landolt-Brnstein
New Series VIII/4

7-40

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

eigenvalues and eigenvectors of the matrix of transfer rate coefficients as described in good texts on
linear algebra (for example 85J1). The methods are referred to as the matrix exponential method and
eigenanalysis. Birchall and James [89B1] described the matrix exponential method and listed the source
code for effective implementation of this method on a desktop computer. The eigenanalysis method has
been implemented by, among others, Killough and Eckerman [84K1], Bertelli [87B1] and Polig [01P1].
A transportable library of state-of-the art numerical routines for eigenvalue problems is available in the
LAPACK Package [00A1]. An advantage of the eigenanalysis approach is that the solution can be
expressed analytically even though it is a numerical approximation. An imaginative hybrid numericalanalytical method has been developed by Leggett and co-workers that is extremely simple, flexible and
highly efficient [93L1]. Some of these methods have been reviewed by Peace [03P1]. With the memory
and speed available in todays desktop computers and the availability of well written solvers, such as
those within ODEPACK, the numerical aspects of compartment modelling are no longer a significant
issue. However, having available a number of different solvers is useful in ensuring an appropriate
approximation to the solution.
The method of calculation of dose is described in Section 7.3 and is encapsulated in Equation 15. The
number of nuclear transformations of the kth chain member in the ith compartment during the period t1 to t2
U ik (t1 ,t 2 ) is given as
t2

U (t1 , t 2 ) = Aik (u ) du
k
i

(32)

t1

where Aik (t ) is obtained as the solution to Equation 30. For protection of workers the integral of Equation
32 is evaluated from time zero (the time of the intake) to 50 years post intake and the integral is denoted
~
as U(50). The distinction between A typically used in the dosimetry of radiopharmaceuticals and U is
that former has no restriction on the upper limit of the integration since it is typically short-lived
radionuclides. The committed equivalent dose coefficient for tissue T, hT, for the worker is thus computed
as
hT =

U
k

k
S

(50) SEE (T S)

(33)

where the outer summation extends over the parent radionuclide and all members of its decay chain and
the inner summation extends over all source regions in which activity may reside. Note that the
compartments in the model must be assigned to the anatomical source regions S in computing US. For
children both the parameters of the biokinetic models and S-factors (or SEE values) change with time.
They must therefore be varied in an effectively continuous manner from the time of intake to age 70
years.
Absorbed fraction data for photons and charged particles are available for six specified ages enabling
sets of S-factors to be generated for newborn, 1, 5, 10, 15-years-olds and adults. At intermediate ages
S-factors can be derived using an interpolation scheme. ICRP has used a linear interpolation in the inverse
total body mass domain. Biokinetic models are specified by ICRP for six standard ages, 3-month-old, 1,
5, 10, 15-year-old, and adult. These models specify the transfer coefficients (ki,j) which determine the
rates at which material is transferred between the different parts of the body (Equation 30). Transfer
coefficients at intermediate ages can be derived using linear interpolation. Numerical methods, which are
characterised by the variable time steps inherent in the method, can easily accommodate time-varying
transfer coefficients and S-factors. For methods which apply an analytical solution the continuous
variation of transfer rates must be modelled discretely using a time-stepping method. Starting at the age of
intake the calculation is advanced by a time-step small enough for the interpolated rates to be held
constant without undue loss of accuracy. The computed activities at the end of one time-step are used as
the initial conditions for the next step. In this way the number of transformations within each step or
interval is computed. S-factors calculated at the beginning or mid-point of a step are taken to apply to the
Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-41

whole interval. The committed dose is then the sum of the doses computed in each time step. In essence
the numerical methods are integrating dose rate (Equation 15) while analytical methods apply Equation
18 to a series of larger time steps. One of the advantages of the analytical methods is in calculations for
adults where rate constants and S-factors are constant; the time step can then be the whole of the period of
interest, e.g. 50 years. Numerical methods are advantageous, however, where the intake is complicated,
perhaps varying in a difficult manner with time.

7.4.2 Sources of dose coefficients


The recommendations of ICRP and other international bodies advance on different fronts at different
rates. This means that it is not always straightforward to identify the most appropriate dose coefficients
for a particular application (e.g. workplace, environment, nuclear medicine). For example, for inhalation
by members of the public, dose coefficients are given for three lung absorption Types (F, M and S) for 31
important elements (Publication 72, 96I1). A default Type is specified for use when the chemical form is
not known. For workers a comprehensive review of lung absorption characteristics for various chemical
forms of radionuclides has not been undertaken since Publication 30 although updated dose coefficients
using the HRTM have been issued [94I1]. The following subsections aim to help the reader identify the
most appropriate dose coefficients at the time of publication of this review.
The results given in ICRP dose compendia take into account the ingrowth of decay products in all
regions of the body following an intake of unit activity of the parent nuclide. They do not take into
account any activity of decay products in the initial intake. Thus doses from any radioactive decay
products present at the time of the intake (perhaps in equilibrium with the parent) may need to be added to
the dose from the parent nuclide.
7.4.2.1 Workers
A compendium of dose coefficients (committed effective dose to 50 years after the intake) for workers for
over 800 nuclides based on the HRTM model (Section 7.2.1), the ICRP Publication 30 model for the GI
tract (Section 7.2.2) and the most recent ICRP systemic models (Section 7.2.5) has been issued in ICRP
Publication 68 [94I1] which implemented the tissue weighting factors in Publication 60 [91I1] (Table
7.1). These results are also given in the Euratom Directive [96E1] and the IAEA Basic Safety Standards
[96I1]. The previous complete source of doses to workers was ICRP Publication 30, published in four
parts between 1979 and 1988 [79I1, 80I1, 80I2, 88I1]. Publication 30 concentrated on giving results as
Annual Limits on Intake (Bq) rather than as dose coefficients.
For workers, inhalation dose coefficients are based on an AMAD of 5 m and specified distributions
of time spent at two levels of exercise sitting and light exercise [94I2].
In support of Publication 68, ICRP has issued Publication 78 [97I2] which contains bioassay
predictions, such as daily urinary excretion and retention in lung, skeleton and whole-body, based on the
same models used in Publication 68 for a limited range of radionuclides. This enables health physicists to
use a consistent set of models in dose assessments. This document fills the role for ICRP Publication 68
that Publication 54 filled for Publication 30. Phipps et al [98P1] provide extended results and fitted
functions for predicting bioassay quantities at times not addressed in the ICRP report. For particle sizes
other than 5 m AMAD, Ishigure et. al. calculated bioassay quantities for 0.1, 0.3, 1, 3 and 10 m and
have uploaded the results onto the National Institute of Radiological Sciences web site [02I4]. Further
details of the methods of monitoring and dose assessment in the workplace are given in Sections 7.4-7.7
and in Chapter 10, Sections 10.3.2. and 10.3.3.
A CD-ROM of dose coefficients for both members of the public and workers has been issued by ICRP
[99I1]. It is consistent with the Publication 68 and it extends the results given in Publications 68 and 72
by giving inhalation dose coefficients for ten particle sizes (0.001, 0.003, 0.01, 0.03, 0.1, 0.3, 1, 3, 5, 10
m AMAD) and ingestion coefficients. Effective doses and equivalent doses for all important tissues for a
Landolt-Brnstein
New Series VIII/4

7-42

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

range of integration periods (1, 7, 30 days, 1, 5, 10, 20, 30 and 45 years) are given, together with the dose
coefficients to age 70 years. The package also contains extensive help files and the text of Publications 68
and 72.
7.4.2.2 Members of the public
Following the Chernobyl accident it was acknowledged that a set of internationally-agreed dose
coefficients was required. Thus, ICRP provided age-specific biokinetic models for selected radionuclides
in Publications 56, 67, 69, 71 and 72 [89I1, 93I1, 95I1, 95I2, 96I1] together with dose coefficients
(committed effective dose to age 70 years) for six age groups: 3-month-old infants, 1-, 5-, 10-, 15-year
old children and adults (Table 7.1). More details about the models themselves are given in Section 7.2.
ICRP Publication 56 [89I1] considered H, C, Sr, Zr, Nb, Ru, I, Cs, Ce, Pu, Am and Np. Dose coefficients
for selected radionuclides were based on the tissue weighting factors of ICRP Publication 26 [77I1] and
the lung model of ICRP Publication 30. These results have now been superseded by other Publications.
ICRP Publication 67 [93I1] considered S, Co, Ni, Zn, Mo, Tc, Ag, Te, Ba, Pb, Po, and Ra, and in addition
revised some of the models given earlier in Publication 56. In particular the model for Sr was revised
substantially and made consistent with the generic model structure used for Ba and Ra. By the time of the
issue of ICRP Publication 67 the tissue weighting factors given in Publication 60 were available thus the
dose coefficients are consistent with the most recent ICRP recommendations. Both equivalent (hT) and
effective (e) dose coefficients were given and the results of ICRP Publication 56 were updated whether or
not the systemic model was updated in Publication 67. Only ingestion was considered as a route of intake.
Publication 69 [95I1] gave a similar range of dose coefficients for radionuclides of Fe, Se, Sb, Th and U.
Following the publication of the model for the Human Respiratory Tract [94I2] ICRP Publication 71
reviewed the lung absorption characteristics of environmental forms of the 31 elements considered in
Publications 56, 67, 69. ICRP Publication 71 also introduced biokinetic models for Ca and Cm. Inhalation
dose coefficients based on the new model were given for all three default Types (F, M and S) and one of
these three was recommended as a default for situations where the chemical form is unknown. Details of
the HRTM model are given in Sction 7.2.1. For members of the public, dose coefficients are based on an
Activity Median Aerodynamic Diameter, AMAD, of 1 m and specified distributions of time spent at
four levels of exercise (sleep, sitting, light exercise and heavy exercise [96I2]). Dose coefficients for
ingestion were not given in Publication 71.
A large compendium of inhalation and ingestion dose coefficients (effective dose only) for members
of the public for over 800 nuclides based on the HRTM model (Section 7.2.1), the ICRP Publication 30
model for the gastrointestinal tract (Section 7.2.2) and the most recent systemic models (Section 7.2.5)
was published in ICRP Publication 72 [96I1]. These results are also given in the Euratom Directive
[96E1] and the IAEA Basic Safety Standards [96I2]. The CD-ROM of dose coefficients (Section 7.4.2.1,
99I1) also contains doses for members of the public consistent with ICRP Publication 72 [99I1]. As noted
above, for 31 elements dose coefficients are given for three default absorption Types (F, M and S), while
for the remaining elements it is assumed that compounds assigned to the Publication 30 classes (D, W and
Y) are categorised as F, M and S respectively; thus for guidance about individual chemical forms of these
elements one must refer to ICRP Publication 30.
7.4.2.3 Embryo and foetus
ICRP has issued dose coefficients for the embryo and foetus in Publication 88 [01I1]. More details on the
biokinetic models used for the dose calculations are given in Section 7.2.7, this subsection therefore
covers only the phantoms used in foetal dosimetry and some aspects of the dosimetry.
The pattern of energy deposition within the foetus is modelled in Publication 88 [01I1] using results
from two separate sets of computer phantoms developed at Oak Ridge National Laboratories, USA
(ORNL). The first is a series for the pregnant female developed by Stabin et al [95S1]; the second is for
the foetus itself, developed by Eckerman [03E1]. Energy-dependent specific absorbed fractions (SAFs)
Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-43

are given for both electrons (including beta particles and positrons) and photons. Due to the small organ
masses and short ranges between organs, for beta radiation the beta spectrum is used and not the mean
energy. In calculating organ and tissue doses for infants, children and adults, electrons have, in most
cases, been assumed to be non-penetrating, i.e. their energy is taken to be absorbed entirely in the organ
or tissue in which they are emitted (exceptions are the skeleton and walled organs). In the case of the
foetus, however, the extremely small size of some tissues can mean that a substantial fraction of electron
energy can be deposited outside the tissue where the electron is emitted [98U1]. Thus, activity in the
foetal thyroid could deliver an electron dose to nearby tissues such as the brain. However, these so-called
cross-fire doses are generally much lower than the doses to the source tissues themselves.
7.4.2.4 Nuclear medicine patients and volunteers in clinical research
ICRP Publication 53 [87I1] presented biokinetic models and best estimates of biokinetic data for some
120 individual radiopharmaceuticals. It included absorbed dose coefficients for organs and effective dose
equivalent coefficients (Publication 30 [79I1] terminology) calculated up to infinity, due to the
administration of short-lived radionuclides for diagnostic or experimental purposes. The calculations used
ICRPs Publication 26 tissue weighting factors [77I1]. Some information on the range of variation to be
expected in pathological states, for adults, children and the foetus were given. The absorbed dose
coefficients are used in clinical diagnostic work for judging the risk associated with the use of specific
radiopharmaceuticals, both for comparison with the possible benefit of the investigation and to help in
giving adequate information to the patient. These estimates also provide guidance to ethics committees
having to decide upon research projects involving the use of radioactive substances in volunteers who
receive no individual benefit from the study. ICRP Publication 53 supplemented ICRP Publication 52,
Protection of the Patient in Nuclear Medicine [88I3].
In Publication 80 [98I1], ICRP provided biokinetic models, absorbed doses to organs and effective
dose coefficients, using the tissue weighting factors of ICRP Publication 60, for 10 new
radiopharmaceuticals. It also provided recalculated dose coefficients for the 19 most frequently used
radiopharmaceuticals from ICRP Publication 53, using ICRP Publication 60 dosimetry. An integrated
index to all radiopharmaceuticals treated in ICRP Publications on nuclear medicine up to 1998 gave a
listing of effective dose coefficients for adults.
Recently Stabin and Siegel [03S1] have used the best current radiation decay data and computer
phantoms to calculate dose coefficients for use in nuclear medicine. Decay data for over 800
radionuclides from the data service at Brookhaven National Laboratory were combined with absorbed
fraction data from a number of currently available mathematical whole body and organ models to
calculate the dose coefficients. Many more (816) radionuclides are considered than in the ICRP
compendia and some alpha emitters are included. New models are also employed, and dose coefficients
for bone and marrow have been updated with recently suggested modifications.

7.4.3 Dose coefficients for selected radionuclides


7.4.3.1 Doses to tissues following intakes of radionuclides
Equivalent doses to tissues following inhalation of 239PuO2 by an occupationally exposed worker are
given in Table 7.12. These doses are calculated using the HRTM [94I1], assuming inhalation Type S and
a 5 m AMAD aerosol, and the biokinetic model for plutonium given in Publication 67 [91I1]. The
highest committed (50 year) doses are to the lung, as the site of entry into the body, and the skeleton
(cells near bone surfaces) and liver, as the main sites of deposition from the blood. ICRP also calculates
the committed effective dose which provides a method for comparing doses, and hence risks, from intakes
of radionuclides with those from external radiation. This is discussed in detail in Chapter 4.
Landolt-Brnstein
New Series VIII/4

7-44

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

Table 7.12. Committed equivalent doses to tissues, weighted committed equivalent doses and committed
effective doses from inhalation by a worker of 239PuO2 (5 m AMAD)a
Tissue weighting
Weighted committed
Organ or tissue
Committed equivalent
factor
equivalent dose [Sv Bq1]
dose [Sv Bq1]
5
Cells near bone surfaces 9.1 10
0.01
9.1 107
7
Colon
0.12
1.7 10
2.0 108
5
Liver
0.05
1.9 10
9.5 107
5
Lungs
0.12
4.7 10
5.6 106
6
Red bone marrow
0.12
4.5 10
5.4 107
7
0.05
Remainder
1.0 108
2.0 10
Skin
0.01
1.5 107
1.5 109
7
Stomach
0.12
1.5 10
1.8 108
6
Gonads
0.20
1.2 10
2.4 107
Committed effective
8.3 106
dose
a Inhalation Type S
Examples of equivalent dose coefficients to tissues from inhalation intakes of 131I, 137Cs and 239Pu by
workers are given in Table 7.13. For an intake of 131I the main dose is to the thyroid, with a dose that is
rather more than 1000 times that to other tissues. For 137Cs, which moves rapidly from the lungs to blood
(Type F) and distributes throughout the body, most tissues receive a very similar dose. In the case of 239Pu
oxide, which has a longer retention time in the lung (Type S) and deposits principally in the liver and
skeleton there is a greater range of doses although the long retention time in the skeleton results in the
highest tissue dose to cells near bone surfaces, as described above.
Table 7.13. Doses to tissues following inhalation by a worker of 131I, 137Cs and 239Pu
Committed equivalent dose [Sv Bq1]
Iodine-131
Tissue
(Type F)
Thyroid
2.1107
Red bone marrow
5.51011
Cells near bone surfaces
6.91011
Colon
5.11011
Lungs
8.11011
Liver
2.41011
Committed effective dose
1.1108
a For inhalation of 5 m AMAD aerosol
b Assumes wR = 20 for particles

Caesium-137
(Type F)

Plutonium-239
(Type S)b

6.3109
6.3109
6.6109
8.1109
6.1109
6.5109
6.7109

1.5107
4.5106
9.1105
1.7107
4.7105
1.9105
8.3106

Some examples of dose coefficients for different radionuclides following intakes by inhalation and
ingestion are shown in Table 7.14. The variation in doses reflects differences in behaviour after intakes by
inhalation or ingestion, variations in distribution and retention in tissues as well as the use of a radiation
weighting factor wR of 20 in the calculation of equivalent dose to tissues from deposited emitters. The
lowest doses are from intakes of tritiated water and the highest doses from inhalation of the emitters
224
Ra, 226Ra and 241Am.

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-45

Table 7.14. Comparison of dose coefficients following inhalation or ingestion of various radionuclides by
a worker.
f1c
Radionuclide
Lung
Dose coefficient [Sv Bq1]
a
Type
Inhalation
Ingestion
3
b
11
H2O

1.0
1.810
1.81011
60
9
Co
M
0.1
3.4109
7.110
90
8
Sr
F
0.3
3.010
2.8108
95
9
Zr
F
0.002
3.010
8.81010
95
9
Nb
M
0.01
1.310
5.81010
106
9
Ru
F
0.05
9.810
7.0109
131
8
I
F
1.0
1.110
2.2108
134
9
Cs
F
1.0
9.610
1.9108
137
9
Cs
F
1.0
6.710
1.3108
144
4
8
Ce
M
510
2.310
5.2109
210
Po
F
0.1
1.1106
6.8107
224
6
Ra
M
0.2
2.410
6.5108
226
5
Ra
M
0.2
1.210
2.8107
232
4
5
Th
M
510
2.910
2.2107
234
6
U
M
0.02
2.110
4.9108
235
6
U
M
0.02
1.810
4.6108
238
6
U
M
0.02
1.610
4.4108
239
5
6
Pu
S
110
8.310
9.0109
241
4
5
Am
M
510
2.710
2.0107
242
4
6
Cm
M
510
3.710
1.2108
a Inhaled materials are classified as Type F, M or S (Fast, Moderate or Slow)
which refer to their rates of absorption to blood from the respiratory tract (Section 7.2.1.2).
AMAD = 5 m.
b Tritiated water is assumed to be completely absorbed from the lungs
c Fractional absorption from the gut
7.4.3.2 Application of dose coefficients in risk calculations
The doses to tissues calculated using the dosimetric models developed by ICRP together with risk
coefficients for fatal cancer recommended by ICRP for the different tissues [91I1] can be used for
assessing the consequences of intakes of radionuclides. An example is given in Table 7.15 for 239Pu
inhaled as the oxide (inhalation Type F, AMAD = 5 m) by a worker.
Table 7.15. Estimation of risk of cancer death following inhalation of 239PuO2 (AMAD 5 m)
Risk for
Tissue
Cancer
Risk coefficient
Sv Bq1
100 kBq inhaled
[cancer deaths Sv1]a
inhaled
5
3
Lungs
Lung
1 in 31
4.710
6.810
5
3
Liver
Liver
1
in 430
1.910
1.210
5
4
Cells near bone surfaces
Bone tumour
1 in 280
9.110
4.010
Red bone marrow
Leukaemia
1 in 550
4.5106 4.0103
Colon
Colon
1 in 8,300
1.7107 6.8103
6
2
Committed effective dose
1 in 26b
8.310
5.010
a Risk coefficients for workers [91I1]
b Based on risks calculated to individual tissues
Landolt-Brnstein
New Series VIII/4

7-46

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

The ICRP dosimetric model for plutonium can be used to estimate the committed equivalent dose to
tissues following an intake by inhalation. By weighting these doses by the appropriate risk coefficients
[91I1], the consequences of an intake of 239PuO2 can be estimated (Table 7.15). The calculations have
been undertaken using the HRTM [94I2] and assuming an AMAD of 5 m (the default value for
workers). The biokinetic model for plutonium given in ICRP Publication 67 (93I1, Figure 7.7) has also
been used.
Following the inhalation of 100 kBq of 239PuO2, for example, the risk of developing lung cancer
would be (4.7105) (6.8103) 105 = 3.2102 i.e. a 1 in 31 risk. The risks of leukaemia, liver and
bone cancer would all be lower, about 1 in 550, 1 in 430 and 1 in 280 respectively. The overall risk of
developing cancer would be about 1 in 26. These risks are clearly a maximum as they are based upon
committed doses and depend upon the full risk to the tissues being expressed. They would therefore apply
only to intakes received early in life. A similar, but approximate calculation could be carried out using the
committed effective dose (8.3106 Sv Bq1) multiplied by the risk coefficient for whole body radiation
exposure recommended by ICRP (5102 Sv1 ) given in Publication 60 [91I1]. For 100 kBq inhaled this
would give an overall risk of 4.15102 (i.e an overall risk of 1 in 24). This is very similar to the value
given in Table 7.15, the difference being mainly due to the fact the ICRP uses rounded values of tissue
weighting factors wT to calculate effective doses. If a more specific calculation was needed for an
individual then it would be necessary to consider the accumulation of dose by the individual over time
(i.e. year on year) and how that risk would be expressed.
Using the same approach the overall risk of developing cancer following either the inhalation or
ingestion of 100 kBq of some of the radionuclides for which dose coefficients are given in Table 7.14 are
given in Table 7.16. There is a significant difference in the risk from intakes of -emitters compared with
/-emitters, reflecting the much higher wR value of 20 for particles. In general the risks of inhalation
intakes are higher than for ingestion because of the lower absorption and faster rate of clearance from the
gut than from the respiratory system. This is not the case for radionuclides such as tritium (as HTO) or
caesium that are readily absorbed from the gut. For the same activity, the risk associated with ingestion of
239
PuO2 is nearly three orders of magnitude less than that following inhalation because of the low
absorption in the gut (f1 = 105). For ingestion of 100 kBq of 131I the risk is about 1 in 38,000, the majority
of the risk of radiation-induced cancers predicted being in the thyroid (more than 99 %). Following
inhalation of the same amount of activity the risk is somewhat lower (1 in 78,000) as only about half of
the activity inhaled is deposited in the respiratory system. Similar considerations apply to the inhalation
and ingestion of 137Cs, although in this case, as the radionuclide distributes throughout body tissues the
risk of cancer will be distributed amongst a number of tissues, with the greatest risks being for leukaemia
and lung cancer. Of the radionuclides considered, 226Ra and 232Th are the most toxic with risks of about 1
in 7.4 and 1 in 10 respectively of developing fatal cancer following inhalation of 100 kBq. At the other
extreme 3H is the least toxic with a risk of less than 1 in a million.

7.5 Internal monitoring


This Section describes the general principles for individual monitoring. Sections 10.3.2 and 10.3.3 in
Chapter 10 give more detailed information on in vivo measurements by whole and partial body counting
as well as by analyses of excreta. This Section is reproduced from ICRP Publication 78, 97I2, paras. 58 76.

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-47

Table 7.16. Comparison of risk of radiation-induced cancer death associated with inhalation or ingestion
of 100 kBq of some radionuclides.
Risk of cancer death
Radionuclide
Lunga Type
f1c
Inhalation
Ingestion
3
b
7
H2O
1.0
1 in 1.410
1 in 1.4107
90
Sr
F
0.3
1 in 9,300
1 in 9,700
95
Zr
F
0.002
1 in 86,000
1 in 170,000
95
Nb
M
0.01
1 in 150,000
1 in 310,000
106
Ru
F
0.05
1 in 22,000
1 in 18,000
131
I
F
1.0
1 in 78,000
1 in 38,000
137
Cs
F
1.0
1 in 37,000
1 in 19,000
144
Ce
M
1 in 9,600
1 in 22,000
3104
210
Po
F
0.1
1 in 310
1 in 870
224
Ra
M
0.2
1 in 64
1 in 3,000
226
Ra
M
0.2
1 in 7.4
1 in 1,200
232
Th
M
1 in 10
1 in 1,300
5104
239
Pu
S
1 in 26
1 in 18,000
1105
241
Am
M
1 in 12
1 in 1,100
5104
242
Cm
M
1 in 46
1 in 15,000
5104
a Inhaled materials are classified as Type F, M or S (Fast, Moderate or Slow)
which refer to their rates of absorption to blood from the respiratory tract (Section 7.2.1.2).
AMAD = 5 m
b Tritiated water is assumed to be completely absorbed from the lungs
c Fractional absorption from the gut

7.5.1 Methods of individual monitoring


The purpose of this Section is to describe briefly the main measurement techniques, their advantages and
their limitations. In most cases, individual monitoring for intakes of radionuclides may be achieved by
body activity measurements, excreta monitoring, air sampling with personal air samplers, or a
combination of these techniques. The choice of measurement technique will be determined by several
factors: the radiation emitted by the radionuclide; the biokinetic behaviour of the contaminant; its
retention in the body taking account of both biological clearance and radioactive decay; the required
frequency of measurements; and the sensitivity, availability, and convenience of the appropriate
measurement facilities.
Routine monitoring programmes usually involve only one type of measurement if adequate sensitivity
can be achieved. For some radionuclides, only one measurement technique is feasible, e.g. urine
monitoring for intakes of tritium. For radionuclides, such as plutonium isotopes, that present difficulties
for both measurement and interpretation, a combination of techniques has to be employed. If different
methods of adequate sensitivity are available, the general order of preference in terms of accuracy of
interpretation is: body activity measurements; excreta analysis; personal air sampling. Results of
monitoring of the working environment (area monitoring) may provide information that assists in
interpreting the results of individual monitoring, e.g. information on particle size, chemical form and
solubility, time of intake. The results of workplace monitoring for air contamination may sometimes be
used to estimate individual intakes. However the interpretation of the results of measurements from air
sampling in terms of intake is not simple and may be misleading. The most common form of
representative sampling is by using fixed samples at a number of selected locations intended to be
reasonably representative of the breathing zone of the worker. When such a method is routinely used for
quantitative determinations of intake, the representativeness of the results should be determined using a
special monitoring programme, often involving personal air samples.
Landolt-Brnstein
New Series VIII/4

7-48

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

Monitoring in relation to a particular task or event may often involve a combination of techniques so
as to make the best possible evaluation of an unusual situation, for example, a programme of both body
activity and excreta measurements and, in some circumstances, personal air sampling. In some cases of
suspected incidents, screening techniques (such as measuring nose blow samples or nasal smears) may be
employed to give a preliminary estimate of the seriousness of the incident. In these cases the regional
deposition for ET1 can be used to confirm that an intake has occurred and to give a rough estimate of the
intake.
7.5.1.1 In Vivo measurements
The IAEA [96I2] has given guidance on the direct measurement of body content of radionuclides. Direct
measurement of body or organ content provides a quick and convenient estimate of activity in the body. It
is feasible only for those radionuclides emitting radiation that can escape from the body. In principle, the
technique can be used for radionuclides that emit: X- or -radiation; positrons, since they can be detected
by measurement of annihilation radiation; energetic -particles that can be detected by measurement of
bremsstrahlung; some -emitters that can be detected by measurement of the characteristic X-rays.
Many facilities for the measurement of radionuclides in the whole body or in regions of the body
consist of one or a number of high efficiency detectors housed in well-shielded, low-background
environments [96I2]. The geometrical configuration of the detectors is arranged to suit the purpose of the
measurement, e.g. the determination of whole-body activity or of activity in a region of the body such as
the thorax or the thyroid. The skull or knees may be used as a suitable site for measurement of
radionuclides in the skeleton.
Care must be taken to remove surface contamination before body activity is measured. For routine
measurements, determination of whole-body content is often adequate for radiological protection
purposes. Total body activity will then consist of systemic activity and activity in the gastrointestinal and
respiratory tracts. However, in special investigations, or in interpretation of unusual measurements, it may
be advantageous to determine the distribution within the body either by profile scanning or by analysis of
the relative response of detectors placed at different positions along the body.
Commonly encountered fission and activation products, such as 131I, 137Cs and 60Co, can be detected
with comparatively simple equipment at levels that are adequate for radiation protection purposes. Such
simple equipment may consist of a single detector, viewing the whole body or a portion of the body, or,
for iodine isotopes, a small detector placed close to the thyroid. The advantage of simple equipment is
that it may be operated at the place of work, thereby avoiding the time required to visit a remote wholebody monitoring facility. Measurements may then be made more frequently so that any unusually large
intake would be recognised soon after it had occurred.
In contrast, high sensitivity techniques are needed for monitoring a few radionuclides at the levels that
are required for protection purposes. Examples are the -emitting radionuclides such as plutonium
isotopes.
Until recently, most body activity measurement facilities, whether high-sensitivity or simple systems,
used thallium-activated sodium iodide detectors. These have the advantage that crystals of large volume
can be manufactured and so provide high efficiency for detection of -rays. Interpretation of a -ray
energy spectrum obtained from a mixture of radionuclides may, however, raise some difficulties. The
components of the spectrum can be resolved by a multiple linear regression analysis technique, but this
requires previous calibration of the detection equipment with standard sources of the required
radionuclides dispersed in a matrix in such a way as to simulate the distribution and attenuation within the
body. The increasing availability of high-efficiency germanium detectors is leading to their use in
situations where workers may be exposed to mixtures of -ray emitting radionuclides. The superior
energy resolving power of these detectors simplifies the interpretation of spectra obtained from complex
mixtures of radionuclides.
The activity present in a wound can be easily detected with conventional - detectors if the
contaminant emits energetic -rays. In the case of contamination with -emitting radionuclides, detection
is more difficult since the low energy X-rays that follow the -decay will be severely attenuated in tissue;
Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-49

this effect is more important the deeper the wound. It is often necessary to localise the active material and
this requires a well-collimated detector. Wound monitors most have an energy discrimination capability if
a good estimate is to be made of contamination with mixtures of radionuclides.
7.5.1.2 Analysis of excreta and other biological materials
In some cases, excreta monitoring may be the only measurement technique for those radionuclides which
have no -ray emission or which have only low energy photon emissions. Excreta monitoring
programmes usually involve analysis of urine, although faecal analysis may be required in some
circumstances, for example where an element is preferentially excreted via faeces or to assess clearance
of Type-S material from the respiratory tract. Other samples may be analysed for specific investigations.
Examples are nose blow or nasal smears as routine screening techniques or blood, in the case of suspected
high level contamination.
The collection of urine samples involves three considerations. Firstly, care must be taken to avoid
adventitious contamination of the sample. Secondly, it is usually necessary to assess the total activity
excreted in urine per unit time from the sample provided. For most routine analyses, a 24 h collection is
preferred but, if this is not feasible, it must be recognised that smaller samples may not be representative.
Tritium is a particular case for which it is usual to take only a small sample and to relate the measured
activity concentration to the concentration in body water. Thirdly, the volume required for analysis
depends upon the sensitivity of the analytical technique. For some radionuclides, adequate sensitivity can
be achieved only by analysis of several days excreta.
The analysis of faecal samples for routine monitoring involves uncertainty in interpretation owing to
daily fluctuations in faecal excretion. Ideally, therefore, collection should be over a period of several
days. However, this may be difficult to achieve in practice and interpretation may need to be based on a
single sample. Faecal monitoring is more often used in special investigation, particularly following a
known or suspected intake by inhalation of Type M or insoluble S compounds. In these circumstances
measurement of the quantity excreted daily may be useful in the evaluation of clearance from the lungs
and in the estimation of intake. Early results may be useful in identifying exposed individuals.
Radionuclides that emit -rays may be determined in biological samples by direct measurement with
scintillation or semiconductor detectors. Analysis of - and -emitting radionuclides requires chemical
separation followed by appropriate measurement techniques. Measurement of so-called total or
activity may occasionally be useful as a simple screening technique, but there is no method that will
determine accurately all the and activity in the sample. The technique may be used in routine
monitoring situations where intakes are expected to be very low compared with annual limits. The results
would not be interpreted quantitatively, but would be used to provide confirmation of satisfactory
conditions, an unusual result indicating the need for further investigation which would include
radiochemical analysis. Total activity measurements may also be useful following a known contamination
event or to identify those samples that merit early attention. Measurements of total or activity cannot
be used in quantitative evaluations of intake or committed effective dose, unless the radionuclide
composition is known.
Measurement of activity in exhaled breath is a useful monitoring technique for some radionuclides
such as 226Ra and 228Th since the decay chains of both these radionuclides include gases which may be
exhaled. It can also be used to monitor 14CO2 formed in vivo from the metabolism of 14C-labelled
compounds.
7.5.1.3 Air sampling
A Personal Air Sampler (PAS) is a portable device specifically designed for the estimation of intake by an
individual worker from a measurement of time-integrated concentration of activity in air in the breathing
zone of the worker. A sampling head containing a filter is worn on the upper torso close to the breathing
zone. Air is drawn through the filter by a calibrated air pump carried by the worker. Ideally, sampling
Landolt-Brnstein
New Series VIII/4

7-50

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

rates would be representative of typical breathing rates for a worker (~1.2 m3 h1). However, sampling
rates of current devices are only about 1/10 of this value. The activity on the filter may be measured at the
end of the sampling period to give an indication of any abnormally high exposures. The filters can then be
retained, bulked over a longer period, and the activity determined by radiochemical separation and high
sensitivity measurement techniques. An estimate of intake during the sampling period can be made by
multiplying the measured integrated air concentration by the volume breathed by the worker during the
period of intake.
There are three important requirements for a PAS device. Firstly, the sampler should collect sufficient
material for the activity corresponding to a significant intake to be measurable in a reasonable counting
time. This will depend mainly on the lowest committed effective dose that the PAS is required to detect.
Typically, in a routine monitoring programme, the requirement will be to detect annual intakes that in
total give rise to committed effective doses greater than 1/10 of the annual dose limit. Secondly, the
volume of air aspirated by the sampler should be sufficient to provide a statistically accurate
representation of the activity concentration in the breathing zone of the worker. PAS monitoring is most
often used for radionuclides such as plutonium, for which a very small number of particles may contain
activities that would correspond to a significant intake. The statistics of sampling small numbers of events
then becomes the critical factor in determining sampling accuracy. Thirdly, the particle collection
characteristics of the sampler should be known. These depend on the aspiration efficiency of the sampling
head and the collection efficiency of the filter. The aspiration efficiency is the ratio of the particle
concentration in the air entering the sampler to that in the ambient air. It is usually close to unity for
particles of aerodynamic diameter less than about 1 m, but the inertia of larger particles will give a
tendency to under- or over-sample according to conditions. Similar effects apply to particles entering the
nose and mouth and are taken into account in the ICRP Human Respiratory Tract Model [94I2] (the
aspiration efficiency of the respiratory tract is termed inhalability).
A PAS does not provide information on particle size. Nevertheless, it is important either to determine
the particle size distribution of the inspirable material or to make realistic assumptions about it, since it
can have a marked effect on deposition fractions in the respiratory tract, and hence on dose estimates.
This is particularly important now that the recommended default AMAD of 5 m is intended to be
realistic rather than conservative in terms of dose estimation [95D1, 97A1]. All samplers are size
selective to a greater or lesser extent, under- or over-sampling at particular particle sizes, and this can
result in errors in intake estimation. The aspiration efficiency of a PAS should therefore be determined to
indicate whether corrections are necessary. An investigation of the aspiration efficiency of a PAS gave
values close to unity up to an aerodynamic diameter of 30 m under workplace conditions [86M1]. It has
been suggested that samplers should be designed to collect the inspirable fraction rather than the total
aerosol [81V1]. Use of such samplers would be acceptable, but would require modification of analysis
procedures, since the ICRP Respiratory Tract Model implicitly assumes that the total aerosol
concentration is known.
Static air samplers (SAS) are commonly used to monitor workplace conditions, but can underestimate
concentrations in air in the breathing zone of a worker, typically by a factor of up to about 10 [80M1].
Nevertheless, if SAS devices are sited appropriately, a comparison of PAS and SAS measurements can be
used to define a PAS:SAS air concentration ratio which can be used in the interpretation of SAS
measurement for dose assessment purposes. It should however be recognised that the use of SAS is a
relatively indirect method for assessing doses, and use of the results to estimate individual dose requires a
careful assessment of exposure conditions and working practices. Apart from their potential use for dose
estimation, SAS devices can also provide useful information on radionuclide composition, and on particle
size if used with a size analyser such as a cascade impactor.

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-51

7.6 Monitoring Programme


(Section 7.6 is reproduced from ICRP Publication 78, 97I2, paras. 81 - 88)

7.6.1 Need for a monitoring programme


ICRP Publication 75 [97I1] recommends that the emphasis in any particular monitoring programme
should be on the formal assessment of doses to those workers who are considered likely to receive
routinely a significant fraction of the relevant dose limit or who work in areas where exposures could be
significant in the event of an accident.
The results of workplace monitoring should give an indication of the likelihood of doses from intakes
exceeding 5 mSv a year. Experience has shown that workers involved in the following operations would
normally require individual monitoring:
handling large quantities of gaseous and volatile materials, e.g. tritium and its compounds in large
scale production processes, in heavy water reactors, and in luminising;
uranium mining and processing and fabrication of uranium and mixed oxide fuels;
processing of plutonium and other transuranic elements;
processing and use of thorium, and
production of large quantities of radionuclides and radiopharmaceuticals.
Selection of the type of monitoring programme depends upon the frequency of contamination of the
workplace. In situations where contamination events are very infrequent, it is unlikely that routine
individual monitoring would be required. Workplace monitoring should be undertaken and the results
used to trigger a programme of individual monitoring in relation to special events. However, for the
processes listed above, if contamination of the workplace occurs frequently, a routine individual
monitoring programme would be appropriate.
For workers who are not routinely employed in areas that are designated as controlled areas in relation
to the control of airborne contamination and who are unlikely to have significant intakes of radionuclides,
routine monitoring of the workplace will usually be sufficient to provide assurance that intakes are
adequately controlled.

7.6.2 Routine monitoring


The required frequency of measurements in a routine monitoring programme depends upon the retention
and excretion of the radionuclide, the sensitivity of the measurement techniques available, and the
acceptable uncertainty in the estimate of intake and committed effective dose. The measurement
technique should be selected so that uncertainties in the measured value are small in relation to the major
source of uncertainty which usually lies in the unknown times of intakes. The frequency of measurements
within a routine monitoring programme should be chosen so as to reduce the uncertainty arising from the
unknown time of intake to an acceptable level.

Landolt-Brnstein
New Series VIII/4

7-52

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

7.6.3 Special or task-related monitoring


Special monitoring refers to monitoring carried out in actual or suspected abnormal situations. Taskrelated monitoring is carried out to provide information about a particular operation. Since both special
and task-related monitoring relate to distinct events, either real or suspected, one of the problems
encountered in interpretation of routine monitoring results does not apply, viz. the time of intake is
known. Furthermore, there may be more information about the physical and chemical form of the
contaminant.

7.6.4 Confirmatory monitoring


One method of confirming that working conditions are satisfactory is to carry out occasional individual
monitoring. Such measurements can be interpreted only qualitatively, but unexpected findings would give
grounds for further investigation. Confirmatory monitoring of this type is most useful for those
radionuclides that are retained in the body for long periods, and occasional measurements provide a check
on the build-up of the activity within the body.

7.6.5 Wound monitoring


When skin is broken, punctured or abraded, radioactive material can penetrate to subcutaneous tissue and
thence be taken up by body fluids. Depending upon the radionuclides and the amount of activity it may be
necessary to undertake a medical investigation and a programme of special monitoring. In these
circumstances, the amount of radioactive material at the site of the wound should be determined taking
into account self-attenuation of the radiation in the foreign material and in tissue, as an aid to decisions on
excision. If an attempt is made to remove material from the wound, measurements should be made of the
removed material and of any activity remaining at the wound site, so as to maintain an activity balance.
Subsequently, a series of measurements should be made to determine uptake to body tissues. These
measurements may consist of in vivo measurements, or urine or faecal excreta monitoring, as appropriate
for the particular radionuclides. If whole-body measurements are made, it may be necessary to shield any
activity remaining at the wound site. Uptake can be assessed from the data given in Section 7.8.
If medical intervention to prevent uptake or enhance excretion is considered, then it should be noted
that any treatment will modify the biokinetic behaviour described by the models given in Section 7.1 and
the data in Section 7.7 cannot be used directly to assess committed effective doses when treatment has
been administered. When therapy is used following an accidental intake, a programme of special
monitoring should be undertaken to follow the distribution and retention of the particular contaminant in
the person, and these data should be used to make a specific assessment of committed effective dose for
that person.

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-53

7.7 Dose Assessment


(Section 7.7 is reproduced from ICRP Publication 78, 97I2, paras. 103 - 109).
Examples for dose estimation from results of in vitro measurements are given in Section 10.3.3.9.

7.7.1 Estimation of intake and dose


For special or task-related monitoring when the time of intake is known, the intake can be estimated from
the measured results using the predicted values of measured quantities as illustrated by Figures 7.15 to
7.25. If only a single measurement is made, the intake can be determined from the measured quantity M
by
Intake =

M
m(t )

(34)

where m(t) is the predicted value at the time of intake t. The intake can be multiplied by the dose
coefficient to give the committed effective dose; this can then be compared with the dose limit or any predetermined investigation level based on dose.
If the measurement indicates that an investigation level has been exceeded, further investigation is
required. The nature of the investigation will depend upon the circumstances and the extent to which the
investigation level is exceeded. The following should be considered:
repeated measurements to confirm or refine the initial evaluation, and
the use of additional monitoring techniques.
If a series of measurements is available, the data in Figures 7.15 to 7.25 provide the time course of the
predicted activity (at least over a period of 10 days). The predicted values can then be scaled to obtain the
best fit to the measured data points. The best fit is usually taken to be that fit which minimizes the sum of
the squares of the residuals, a residual being defined as the number of standard deviations separating a
measurement from the fitted curve. The intake is then equal to the value by which the predicted values are
scaled.
For routine monitoring, it is assumed that intake took place in the middle of the monitoring interval of
T days. For a given measured quantity M obtained at the end of the monitoring interval, the intake is
Intake =

M
m(T 2 )

(35)

and the dose from intake in the monitoring interval is obtained by multiplying the intake by the dose
coefficient. The dose or intake can be compared with the dose limit or of the activity corresponding to
that limit. Alternatively, the dose or intake can be compared with pre-determined investigation levels.
An intake in a preceding monitoring interval may influence the actual measurement result obtained. If
more than about 10 % of the actual measured quantity may be attributed to intakes in previous intervals,
for which intake and dose have already been assessed, a correction should be made. For a series of
measurements in a routine monitoring programme, the following procedure may be observed:

Landolt-Brnstein
New Series VIII/4

7-54

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

determine the magnitude of the intake in the first monitoring interval;


predict from the graphs in Section 7.8 the contribution to the subsequent measurement from this
intake;
subtract this contribution from all subsequent data, and
repeat above for the next monitoring interval.
Alternative techniques for assessing committed effective dose from a series of measurement values
are described in the literature, e.g. [96P1].
If a measured value in a routine monitoring programme exceeds a pre-determined investigation level,
further investigation is required. The nature of the investigation will depend upon the circumstances and
the extent to which the investigation level is exceeded.
The following should be considered:

repeated measurements to confirm or refine the initial estimate;


the use of additional monitoring techniques;
review of the working conditions and the circumstances of the exposure;
if default parameter values were used in the original assessment, investigation of the particle size and
chemical form of the actual contaminant and selection of more appropriate values, if necessary, and
in cases of substantial intakes, removal of the contaminated person from work with radioactive
materials and investigation of the actual retention and excretion characteristics, in order to refine the
dose assessment.

7.7.2 Control of worker doses


The limit on the annual effective dose to a worker applies to the sum of the effective doses from external
exposure and committed effective dose from intakes of radionuclides. For practical purposes, the total
effective dose ET can be calculated from the formula:
ET = H p (d ) +

e
j

j ,inh

(50) I j ,inh +

j ,ing

(50) I j ,ing

(36)

where Hp(d) is the personal dose equivalent at a depth d in the body, normally 10 mm for penetrating
radiation, ej.inh(50) is the committed effective dose per unit activity intake by inhalation from radionuclide
j, integrated over 50 years, Ij.inh is the intake of radionuclide j, by inhalation, ej,ing(50) is the committed
effective dose per unit activity intake by ingestion from radionuclide j, integrated over 50 years , and Ij,ing
is the intake of radionuclide j by ingestion.
Strictly, personal dose equivalent is an operational quantity measured in the workplace using personal
dosemeters, whereas the committed effective dose quantities are calculated using measurements of other
parameters (e.g. air concentrations) in the workplace. However, for practical purposes the two kinds of
quantity can be combined in the assessment of the total effective dose.
In the assessment of committed effective doses from internal radionuclides it is often helpful to work
in terms of the secondary quantities: Annual Limit on Intake (ALI, Bq); and Derived Air Concentration
(DAC, Bqm3). The ALI is the intake which would lead to a committed effective dose of 20 mSv (the
average annual limit on effective dose).
ALI =

0.02
e(50)

(37)

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-55

The DAC is the activity concentration in air which would lead to an intake of one ALI assuming a
standard breathing rate (1.2 m3 h1) and annual working hours (2,000).
DAC =

ALI
1.2 2000

(38)

These values should not be seen as limits in the way that the 5-year-averaged effective dose is limited,
but rather as helpful guides to whether the limits are likely to be approached or exceeded.

7.8 Monitoring data for radionuclides


In this Section illustrative graphs of predicted values of measured quantities (whole-body retention,
specific organ retention, daily urinary or faecal excretion) are given in Figs 7.15 to 7.25 as a function of
time following a single intake by inhalation, ingestion and injection. The data for the following
radionuclides are included: 3H, 60Co, 90Sr, 106Ru, 131I, 134Cs, 137Cs, 144Ce, 234, 235, 238U, 239, 240Pu, and 241Am.
For inhalation, results are generally given for a single lung clearance Type which is representative of
chemical forms present in the workplace (see Table 7.4). For tritium, a graph for inhalation of tritiated
water is given which is treated as Class SR-2. In the case of 239, 240Pu and 241Am graphs for both Type M
and Type S forms are given. For ingestion, f1 values recommended for materials in the workplace are
applied (see Table 7.6). For direct entry into the blood the graphs are applicable to soluble (transportable)
forms of radionuclides that have been directly injected into the bloodstream or have entered the body by
inhalation, ingestion or through skin/wound contamination.
The Human Respiratory Tract Model in ICRP Publication 66 [94I2] was used to calculate particle
deposition and respiratory tract clearance of the deposited particles. The subject exposed to the aerosols
was the ICRP reference worker doing light work: defined as light exercise with the ventilation rate of
1.5 m3 h1 for 5.5 h + sitting with the rate of 0.54 m3 h1 for 2.5 h. The following ICRP default values
[94I2] for the physical characteristics of the radioactive aerosols were used.

Activity Median Aerodynamic Diameter (AMAD) = 5 m


geometric standard deviation of particle size = 2.5
particle density = 3 g cm3
particle shape factor = 1.5

The GI tract model in ICRP Publication 30 [79I1] was used. The rate constant B for the absorption of
the materials from the small intestine to the blood was obtained from the value of f1, the fraction of
materials absorbed into blood from the small intestine, using the equation:

B = f1SI / (1 f1 )

(39)

where SI is the rate constant of material transfer from the small intestine to the upper large intestine. If f1
value is 1, 0.99 was taken for calculation, which is in line with the ICRP publications.
The latest ICRP biokinetic models at 2003 were used, which are given in the ICRP publications listed
in Table 7.1.
In the graphs of Figs 7.15 to 7.25 body or organ retention for day 1 means the content at the end of
day 1 etc. For excreted activities, the value at day 1 represents the activity excreted during the first day
after intake, corrected for radioactive decay to the end of day 1. One exception to this is for the intake of
tritiated water; the activity concentration in urine was calculated by dividing the whole body activity at
the time of sampling by the volume of body water, 42 litres. In the context of in vivo measurements, the
following definitions are relevant. Whole-body retention is the sum of systemic material (including that in
the urinary bladder) and material retained within the respiratory and gastrointestinal tracts. The lung
retention is taken to be the sum of the contents of the thoracic lymph nodes and the bronchial,
bronchiolar, and alveolar-interstitial regions.
Landolt-Brnstein
New Series VIII/4

7-56

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

Hydrogen-3 (half-life = 12.3 y)


Possible chemical forms of 3H to which workers are exposed include tritium gas (HT), tritiated water
(HTO) and organically bound tritium (OBT) [89I1].
Tritium emits low energy -particles (0.0057 MeV in average) with 100 % yield and is readily
detected by liquid scintillation counting of a urine sample. A typical detection limit readily achievable in
monitoring programme is 100 Bq/l for urine samples [97I2]. Since the activity concentration of HTO in
urine is assumed to be equal to that in body water, the analysis of HTO in a urine sample is used to give
activity concentration in body water at the time of sample collection.
0

10
H-3 (Water)

H-3 (OBT), f1=1

10-1

Fraction of ingested activity [-]

Fraction of inhaled/ingested/injected activity [-]

100

10-2
10-3
10-4
10-5
10-6
10-7
10-8
100

Whole body
Concentration
in urine
101

102
103
Time after intake [d]

104

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Whole body
Urine
0

10

10
10
Time after intake [d]

10

Fig. 7.15. Predicted whole-body retention, daily urinary excretion or concentration in urine of 3H as a function of
time following acute intake of unit activity of 3H via a all intake routes for tritiated water, b ingestion of organically
bound tritium (OBT), the f1 value of which is 1.0.

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-57

Cobalt-60 (half-life = 5.27 y)


Insoluble compounds of cobalt, e.g. oxides, hydroxides, halides and nitrates are assigned to Type S
(f1=0.05 for workers, 0.01 for adult members of the public) and all other compounds to Type M (f1=0.1)
by ICRP [94I1].
Cobalt-60 emits two high-energy -rays (1.173, 1.332 MeV) per disintegration, which are highly
penetrable radiations and therefore readily detected by photon detectors positioned outside the body. A
typical detection limit readily achievable in monitoring programme is 50 Bq of 60Co in the whole body
and 100 Bq in the lungs [97I2]. Gamma-ray spectrometry on biological samples permits detection of 1
Bq/l of 60Co in urine and 1 Bq per sample of faeces [97I2].
0

100

10
Co-60, Type M, 5 m

Co-60, f 1=0.1

Fraction of ingested activity [-]

Fraction of inhaled activity [-]

10-1
10-2
10-3
10-4
10-5
10-6

Whole body
Urine
Faeces
Lungs

10-7

10-8
100

101

102
103
Time after intake [d]

104

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Whole body
Urine
Faeces
0

10

10
10
Time after intake [d]

10

10

Fraction of injected activity [-]

Co-60

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Whole body
Urine
Faeces
0

Landolt-Brnstein
New Series VIII/4

10

10
10
Time after intake [d]

10

Fig. 7.16. Predicted whole-body/lung retention, or daily


urinary/faecal excretion of 60Co as a function of time
following acute intake of unit activity of 60Co via a
inhalation of particulate aerosols of Type M compounds
with the AMAD of 5 m, b ingestion of compounds
whose f1 value is assumed to be 0.1, c injection of 60Co
in soluble forms.

7-58

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

Strontium-90 (half-life = 29.1 y)


All compounds of strontium possibly present in the work place, except for strontium titanate (SrTiO3), are
assigned to Type F (f1=0.3) by ICRP [94I1]. Strontium titanate is assigned to Type S (f1=0.01) [94I1].
Strontium-90 emits -particles (0.20 MeV in average) with 100 % yield but does not emit energetic
photons. Internally deposited Sr-90 is therefore measured by counting of a urine sample following
chemical separations. A typical detection limit readily achievable in monitoring programme is 1 Bq/l of
90
Sr in urine [97I2].
The decay product of 90Sr, 90Y is radioactive (half-life = 64 h), which emits high-energy -particles
(0.99 MeV in average) with 100 % yield per disintegration of 90Sr. Strontium-90/yttrium-90 in the body
can sometimes be measured by photon detectors positioned outside the body via the bremsstrahlung
produced, though the minimum detectable activities are relatively high [99I2].
10

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

10

Sr-90, f 1=0.3

Fraction of ingested activity [-]

Fraction of inhaled activity [-]

Sr-90, Type F, 5 m
10

Whole body
Urine
Faeces
Skeleton
0

10

10
10
Time after intake [d]

10

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Whole body
Urine
Faeces
Skeleton
0

10

10
10
Time after intake [d]

10

Fraction of injected activity [-]

Sr-90

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Whole body
Urine
Faeces
Skeleton
0

10

10
10
Time after intake [d]

10

Fig. 7.17. Predicted whole-body/skeletal retention, or


daily urinary/faecal excretion of 90Sr as a function of
time following acute intake of unit activity of 90Sr via
a inhalation of particulate aerosols of Type F compounds with the AMAD of 5 m, b ingestion of
compounds whose f1 value is assumed to be 0.3,
c injection of 90Sr in soluble forms.

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-59

Ruthenium-106 (half-life = 1.01 y)


Oxides and hydroxides of ruthenium are assigned to Type S (f1=0.05), halides to Type M (f1=0.05) and all
other compounds to Type F (f1=0.05) by ICRP in Publication 68 for workers [94I1].
Though 106Ru does not emit energetic photons, the radioactive decay product 106Rh (half-life = 30 s)
emits -rays of 0.512 MeV (20.6 % per disintegration of 106Ru), 0.622 MeV (9.8 %) and 1.050 MeV
(1.5 %). They are penetrable radiations and therefore readily detected by photon detectors positioned
outside the body. A typical detection limit readily achievable in monitoring programme is 200 Bq of
106
Ru in the whole body [97I2]. Gamma-ray spectrometry on biological samples permits detection of 5
Bq/l of 106Ru in urine [97I2].
0

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

10

10
Ru-106, Type M, 5 m

Whole body
Urine
Faeces
0

10

10
10
Time after intake [d]

Ru-106, f1=0.05

Fraction of ingested activity [-]

Fraction of inhaled activity [-]

10

10

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Whole body
Urine
Faeces
0

10

10
10
Time after intake [d]

10

Fraction of injected activity [-]

Ru-106

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Whole body
Urine
Faeces
0

Landolt-Brnstein
New Series VIII/4

10

10
10
Time after intake [d]

10

Fig. 7.18. Predicted whole-body retention, or daily


urinary/faecal excretion of 106Ru as a function of time
following acute intake of unit activity of 106Ru via
a inhalation of particulate aerosols of Type M
compounds with the AMAD of 5 m, b ingestion of
compounds whose f1 value is assumed to be 0.05,
c injection of 106Ru in soluble forms.

7-60

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

Iodine-131 (half-life = 8.04 d)


Elemental iodine vapour is assigned to Class SR-1 (10 % deposition in ET1, 40 % in ET2, 50 % in BB),
with Type F clearance [94I1, 95I2]. Methyl iodide gas is assigned to Class SR-1 (70 % deposition in ET2
and the lungs), with Type V clearance [95I2]. For workers particulate aerosols of iodine compounds are
all assigned to Type F (f1=1.0) [94I1].
Iodine-131 emits -rays of 0.284 MeV (6.1 % yield), 0.364 MeV (81.2 %), and 0.637 MeV (7.3 %).
The principal -ray emissions at 0.364 MeV are used for measurement of 131I by photon detectors
positioned just outside the thyroid, in which iodine is highly accumulated. A typical detection limit
readily achievable in monitoring programme is 100 Bq of 131I in the thyroid [97I2]. Gamma-ray
spectrometry on biological samples permits detection of 1 Bq/l of 131I in urine [97I2].
10

10

I-131, f1=1

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

10

Fraction of ingested activity [-]

Fraction of inhaled activity [-]

I-131, Vapour
10

Thyroid
Urine
Faeces
0

10

10
10
Time after intake [d]

10

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Thyroid
Urine
Faeces
0

10

10
10
Time after intake [d]

10

Fraction of injected activity [-]

I-131

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Thyroid
Urine
Faeces

10

10
10
Time after intake [d]

10

Fig. 7.19. Predicted thyroid retention, or daily


urinary/faecal excretion of 131I as a function of time
following acute intake of unit activity of 131I via
a inhalation of iodine vapour, b ingestion of compounds
whose f1 value is assumed to be 1.0 (0.99 was taken in
this publication for computational reasons [95I2]),
c injection of 131I in soluble forms.

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-61

Caesium-134 (half-life = 2.06 y)


All compounds of caesium possibly present in work place are assigned to Type F (f1=1.0) by ICRP [94I1]
although it is recognised that other forms may be present in the environment [95I2].
Caesium-134 emits -rays of 0.563 MeV (8.4 % yield), 0.569 MeV (15.4 %), 0.605 MeV (97.6 %),
0.796 MeV (85.4 %) and 0.802 MeV (8.7 %), which are penetrable radiations and therefore readily
detected by photon detectors positioned outside the body. A typical detection limit readily achievable in
monitoring programme is 50 Bq of 134Cs in the whole body [97I2]. Gamma-ray spectrometry on
biological samples permits detection of 1 Bq/l of 134Cs in urine [97I2].
0

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

10

10

Cs-134, Type F, 5 m

Whole body
Urine
Faeces
0

10

10
10
Time after intake [d]

10

Cs-134, f 1=1

Fraction of ingested activity [-]

Fraction of inhaled activity [-]

10

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Whole body
Urine
Faeces
0

10

10
10
Time after intake [d]

10

Fraction of injected activity [-]

Cs-134

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Whole body
Urine
Faeces
0

Landolt-Brnstein
New Series VIII/4

10

10
10
Time after intake [d]

10

Fig. 7.20. Predicted whole-body retention, or daily


urinary/faecal excretion of 134Cs as a function of time
following acute intake of unit activity of 134Cs via
a inhalation of particulate aerosols of Type F compounds
with the AMAD of 5 m, b ingestion of compounds
whose f1 value is assumed to be 1.0 (0.99 was taken in
this publication for computational reasons [95I2]),
c injection of 134Cs in soluble forms.

7-62

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

Caesium-137 (half-life = 30.0 y)


All compounds of caesium possibly present in work place are assigned to Type F (f1=1.0) by ICRP [94I1]
although it is recognised that other forms may be present in the environment [95I2].
Though 137Cs does not emit energetic photons, the radioactive decay product 137mBa (half-life =
2.55 min) emits -rays of 0.662 MeV (85.0 % per disintegration of 137Cs), which are penetrable radiations
and therefore readily detected by photon detectors positioned outside the body. A typical detection limit
readily achievable in monitoring programme is 50 Bq of 137Cs in the whole body [97I2]. Gamma-ray
spectrometry on biological samples permits detection of 1 Bq/l of 137Cs in urine [97I2].
0

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

10

10

Cs-137, Type F, 5 m

Whole body
Urine
Faeces
0

10

10
10
Time after intake [d]

10

Cs-137, f 1=1

Fraction of ingested activity [-]

Fraction of inhaled activity [-]

10

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Whole body
Urine
Faeces
0

10

10
10
Time after intake [d]

10

Fraction of injected activity [-]

Cs-137

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Whole body
Urine
Faeces
0

10

10
10
Time after intake [d]

10

Fig. 7.21. Predicted whole-body retention, or daily


urinary/faecal excretion of 137Cs as a function of time
following acute intake of unit activity of 137Cs via
a inhalation of particulate aerosols of Type F compounds
with the AMAD of 5 m, b ingestion of compounds
whose f1 value is assumed to be 1.0 (0.99 was taken in
this publication for computational reasons [95I2]),
c injection of 137Cs in soluble forms.

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-63

Cerium-144 (half-life = 284 d)


Oxides, hydroxides and fluorides of cerium are assigned to Type S (f1=0.0005) and all other compounds
to Type M (f1=0.0005) by ICRP in Publication 68 [94I1].
Cerium-144 emits -rays of 0.080 MeV (1.6 % yield) and 0.134 MeV (10.8 %). The radioactive decay
product of 144Ce, 144Pr emits -rays of 0.697 MeV (1.5 % per disintegration of 144Ce). Because of their low
abundances, detection limits of in vivo counting for 144Ce are relatively high; a typical detection limit that
can be readily achieved is 10 kBq of 144Ce in the whole body [88I1]. Detection limits lower than this
value are required for routine monitoring. Urine monitoring is not recommended, because cerium in the
body is tenaciously retained and hardly excreted (biological half-life = 3500 d [79I1]).
10

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

10

Ce-144, f1=0.0005

Fraction of ingested activity [-]

Fraction of inhaled activity [-]

Ce-144, Type M, 5 m
10

Whole body
Urine
Faeces
0

10

10
10
Time after intake [d]

10

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Whole body
Faeces

10

10
10
Time after intake [d]

10

Fraction of injected activity [-]

Ce-144

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Whole body
Urine
Faeces
0

Landolt-Brnstein
New Series VIII/4

10

10
10
Time after intake [d]

10

Fig. 7.22. Predicted whole-body retention, or daily


urinary/faecal excretion of 144Ce as a function of time
following acute intake of unit activity of 144Ce via
a inhalation of particulate aerosols of Type M compounds with the AMAD of 5 m, b ingestion of compounds whose f1 value is assumed to be 0.0005,
c injection of 144Ce in soluble forms.

7-64

7 Internal dosimetry of radionuclides

[Ref. p. 7-68

Uranium-234 (half-life = 2.44105 y), -235 (half-life = 7.04108 y), -238 (half-life
= 4.47109 y)
In Publication 68 [94I1], ICRP assigned most hexavalent compounds of uranium, e.g. UF6, UO2F2 and
UO2(NO3)2 to Type F (f1 = 0.02), less soluble compounds, e.g. UO3, UF4, UCl4 and most other hexavalent
compounds to Type M (f1 = 0.02) and highly insoluble compounds, e.g. UO2 and U3O8 to Type S
(f1 = 0.002).
Principal isotopes of uranium (234U, 235U, 238U) are -emitting radionuclides and do not emit energetic
photons except for 235U. Internally deposited uranium-isotopes are therefore measured by -spectrometry
on biological samples following radiochemical separation. A typical detection limit is 10 mBq/l in urine
and 10 mBq in faeces [97I2]. Uranium-235 emits -rays of 0.144 MeV (10.5 % yield), 0.186 MeV
(54.0 %) and 0.205 MeV (4.7 %). They are used for lung counting of 235U. A typical detection limit is 200
Bq [97I2]. For routine monitoring, the detection limits for -spectrometry are adequate, but those for lung
counting would not permit detection of intakes at annually limited levels [97I2].
10

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

10

U-234/235/238, f 1=0.02

Fraction of ingested activity [-]

Fraction of inhaled activity [-]

U-234/235/238, Type M, 5 m
10

Lungs
Urine
Faeces
Skeleton
0

10

10
10
Time after intake [d]

10

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Whole body
Urine
Faeces
Skeleton

10

10
10
Time after intake [d]

10

Fraction of injected activity [-]

U-234/235/238

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Whole body
Urine
Faeces
Skeleton
0

10

10
10
Time after intake [d]

10

Fig. 7.23. Predicted whole-body/lung/skeletal retention,


or daily urinary/faecal excretion of 234/235/238U as a
function of time following acute intake of unit activity of
234/235/238
U via a inhalation of particulate aerosols of
Type M compounds with the AMAD of 5 m,
b ingestion of compounds whose f1 value is assumed to
be 0.02, c injection of 234/235/238U in soluble forms.

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]

7 Internal dosimetry of radionuclides

7-65

Plutonium-239 (half-life = 2.41104 y), -240 (half-life = 6.54103 y)


In Publication 68 [94I1], ICRP assigned insoluble oxides of plutonium, e.g. high-fired PuO2, a common
chemical form in nuclear industry, to Type S (f1=0.00001) and all other compounds to Type M
(f1=0.0005). Among Type M compounds, f1-value of nitrates is assumed to be 0.0001 [94I1].
Plutonium-239/240 are -emitting radionuclides and do not emit energetic photons. Internally
deposited 239/240Pu are therefore measured by -spectrometry on biological samples following
radiochemical separation. A typical detection limit is 1 mBq/l in urine and 1 mBq in faeces [97I2].
Emission of low energy characteristic X-rays (0.014 - 0.020 MeV) are used for lung counting of 239/240Pu.
A typical detection limit is 2 kBq, which is not adequate for routine monitoring [97I2].
100

10

Pu-239/240, Type S, 5 m

Pu-239/240, Type M, 5 m

10-2

Fraction of inhaled activity [-]

Fraction of inhaled activity [-]

10-1

10-3
10-4
10-5
10-6
10-7

Lungs
Urine
Faeces
Skeleton

10-8
100

101

10

102
103
Time after intake [d]

104

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

10

Lungs
Urine
Faeces
Skeleton

10

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Skeleton
Urine
Faeces

Landolt-Brnstein
New Series VIII/4

10

10
10
Time after intake [d]

10

10

Pu-239/240, f1=0.0001

Fraction of ingested activity [-]

Fraction of ingested activity [-]

-1

Pu-239/240, f1=0.0005
10

10
10
Time after intake [d]

10

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Skeleton
Urine
Faeces

10

10
10
Time after intake [d]

7-66

7 Internal dosimetry of radionuclides


10

10

[Ref. p. 7-68

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Fraction of injected activity [-]

Fraction of ingested activity [-]

Pu-239/240, f 1=0.00001
10

Skeleton
Urine
Faeces

10

10
10
Time after intake [d]

10

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Pu-239/240

Skeleton
Urine
Faeces
0

10

10
10
Time after intake [d]

10

Fig. 7.24. Predicted lung/skeletal retention, or daily urinary/faecal excretion of 239/240Pu as a function of time
following acute intake of unit activity of 239/240Pu via a inhalation of particulate aerosols with the AMAD of 5 m of
Type M compounds, b Type S compounds, c ingestion of compounds whose f1 value is assumed to be 0.0005, d
0.0001, e 0.00001, f injection of 238/2390Pu in soluble forms.

Am-241 (half-life = 4.32102 y)


All compounds of americium possibly present in work place are assigned to Type M (f1=0.0005) by ICRP
[94I1]. Based on several experimental results, ICRP considers that the trace contaminant 241Am that has
grown from 241Pu in matrices of nuclear fuels behaves similarly to the bulk materials [95I2]. For this
reason, Type S as well as Type M is taken in this publication.
Americium-241 is -emitting radionuclide accompanied with low-energy (0.060 MeV) -ray emission
with 35.7 % yield. Internally deposited 241Am can be measured by both indirect and direct methods. A
typical detection limit of -spectrometry following radiochemical separation is 1 mBq/l in urine and
1 mBq in faeces [97I2]. These detection limits are adequate for both special and routine monitoring.
Typical detection limits of in vivo measurements are 20 Bq for lungs and 20 Bq for skeleton. These
detection limits are not necessarily adequate for routine monitoring [97I2].

Landolt-Brnstein
New Series VIII/4

Ref. p. 7-68]
10

7 Internal dosimetry of radionuclides

10

7-67

Am-241, Type S, 5 m

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

10

Fraction of inhaled activity [-]

Fraction of inhaled activity [-]

Am-241, Type M, 5 m
10

Lungs
Urine
Faeces
Skeleton
0

10

10
10
Time after intake [d]

10

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

10

Lungs
Urine
Faeces
Skeleton
0

10

10
10
Time after intake [d]

10

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Fraction of injected activity [-]

Fraction of ingested activity [-]

Am-241, f1=0.0005
10

Skeleton
Urine
Faeces

10

10
10
Time after intake [d]

10

10

-1

10

-2

10

-3

10

-4

10

-5

10

-6

10

-7

10

-8

10

Am-241

Skeleton
Urine
Faeces
0

10

10
10
Time after intake [d]

Fig. 7.25. Predicted lung/skeletal retention, or daily urinary/faecal excretion of 241Am as a function of time following
acute intake of unit activity of 241Am via a inhalation of particulate aerosols with the AMAD of 5 m of Type M
compounds, b Type S compounds, c ingestion of compounds whose f1 value is assumed to be 0.0005, d injection of
241
Am in soluble forms.

Landolt-Brnstein
New Series VIII/4

7-68

7 Internal dosimetry of radionuclides

7.9 References
10B1 Bateman, H.: Proc. Cambridge Philos. Soc. 16 (1910) 423.
56M1 Marinelli, L.D., Miller, C.E.: Gamma Ray Activity of Contemporary Man. Science 124 (1956)
122.
59I1 ICRP: Recommendations of the International Commission on Radiological Protection. Report of
committee II on permissible dose for internal radiation. ICRP Publication 2. Oxford: Pergamon
Press, 1959.
68B1 Berger, M.J.: MIRD Pamphlet No. 2. Energy Deposition in Water by Photons from Point
Isotropic Sources; J Nucl. Med. 9: Suppl. No. 1 (1968) 15-25.
69S1 Snyder, W.S., Ford, M.R., Warner, G.G., Fisher, H.L.: Estimates of absorbed fractions for
monoenergetic photon sources uniformly distributed in various organs of a heterogeneous
phantom. J. Nucl. Med. 10 (suppl 2) (1969) 5.
71B1 Berger, M.J.: Distribution of absorbed dose around point sources of electrons and Beta particles in
water and other media. J. Nucl. Med. 12 (suppl 5) (1971) 5.
71G1 Gear, C.W.: Numerical initial value problems in ordinary differential equations, Englewood
Cliffs, N.J.: Prentice Hall, 1971.
73S1 Spencer, L.V., Simmons, G.L.: Improved Moment Method Calculations of Gamma-Ray
Transport: Application to Point Isotopic Sources in Water; Nucl. Sci. Eng. 50 (1973) 20-31.
74H1 Hindmarsh, A.C.: GEAR: Ordindary differential equations system solver, Lawrence Livermore
National Laboratory Report UCID-30001, Rev. 3, 1974.
74S1 Skrable, K., French, C., Chabot, G., Major, A.: General equation for the kinetics of linear first
order phenomena. Health Phys. 27 (1974) 155.
75I1 ICRP. Report on the Task Group on Reference Man. ICRP Publication 23. Oxford: Pergamon
Press, 1975.
76L1 Loevinger, R., Berman, M.A.: Revised schema for calculating the absorbed dose from
biologically distributed radionuclides: MIRD Pamphlet No 1, Revised. Soc. Nucl. Medicine, NY,
1976.
77I1 ICRP: Recommendations of the International Commission on Radiological Protection. ICRP
Publication 26. Ann. ICRP 1 (3) (1977); reprinted (with additions) in 1977.
77U1 UNSCEAR: United Nations Scientific Committee on the Effect of Atomic Radiation Sources and
Effect of Ionizing Radiation. 1977 Report to the General Assembly, with Annexes. Annex C
Radioactive Contamination due to Nuclear Explosions, 1977.
78S1 Snyder, W.S., Ford, M.R., Warner, G.G.: Medical Internal Radiation Dose Committee (MIRD)
Pamphlet No. 5 (revised). New York, USA: Society of Nuclear Medicine, 1978.
79I1 ICRP: Limits for intakes of radionuclides by workers. ICRP Publication 30, Part 1. Oxford:
Pergamon Press, 1979.
80I1 ICRP: Limits for intakes of radionuclides by workers. ICRP Publication 30, Part 2. Oxford:
Pergamon Press, 1980.
80I2 ICRP: Limits for intakes of radionuclides by workers. ICRP Publication 30, Part 3. Oxford:
Pergamon Press, 1980.
80I3 ICRP: Biological effects of inhaled radionuclides. ICRP Publication 31. Ann. ICRP 4 (1/2).
Oxford: Pergamon Press, 1980.
80M1 Marshall, M., Stevens, D.C.: The Purposes, Methods and Accuracy of Sampling for Airborne
Particulate Radioactive Materials; Health Phys. 39 (1980) 409-423.
80W1 Watson, S.B. and Ford, M.R. A Users Manual to the ICRP Code- A Series of Computer
Programs to Perform Dosimetric Calculations for the ICRP Committee 2 Report, Oak Ridge
National Laboratory Report ORNL/TM-6980 (1980).
81V1 Vincent, J.H., Armbruster, I.: On the quantitative definition of the inhalability of airborne dust.
Ann. Occup. Hyg. 24 (1981) 245.
83H1 Hindmarsh, A.C.: ODEPACK, a systematized collection of ODE solvers, in: Scientific
Computing, Stepleman, R.S., et al. (eds.), Amsterdam: North-Holland, 1983, p. 55.
Landolt-Brnstein
New Series VIII/4

7 Internal dosimetry of radionuclides


83I1
83S1
83S2

84K1
84L1
85J1
86I1
86M1
87B1
87C1
87I1
87S1
88B1
88I1
88I2
88I3
88N1
88Z1
89B1
89I1
89M1
89T1
91I1
91I2
93I1

7-69

ICRP.: Radionuclide transformations: Energy and intensity of emissions. ICRP Publication 38.
Ann. ICRP 11-13. Oxford: Pergamon Press, 1983.
Stather, J.W., Greenhalgh, J.R.: The metabolism of iodine in children and adults. NRPB-R140,
Chilton, 1983.
Stieve, F.E.: Exchange and transfer mechanisms of radioactive compounds between the mother
and the developing off-spring in utero Review of the experimental literature. In: Streffer, C.,
Patrick, G. (eds). Effects of prenatal irradiation with special emphasis on late effects. EUR 8067
EU, Commission of the European Communities, 1983, p. 159.
Killough., G.G., Eckerman, K.F.: A Conversational Eigenanalysis Program for Solving
Differential Equations. Proceedings of the 17th Midyear Topical Symposium of the Health Physic
Society, Ed (R.L. Kathren, D.P. Higby, and M.A. McKinney) (1984).
Leggett, R.W., Dunning jr., D.E., Eckerman, K.F.: Modelling the behaviour of chains of
radionuclides inside the body. Radiat. Prot. Dosim. 9 (2) (1984) 77.
Jacques, J.A.: Compartmental analysis in biology and medicine. Ann Arbor: The University of
Michigan Press, 1985.
ICRP: The metabolism of plutonium and related elements. ICRP Publication 48. Ann. ICRP 16
(2/3). Oxford: Pergamon Press, 1986.
Mark, D., Vincent, J.H., Stevens, D.C., Marshall, M.: Investigation of the entry characteristics of
dust samplers of a type used in the British nuclear industry. Atmos. Environ. 20 (1986) 2389.
Bertelli, L., Lipsztein, J.L.: A mathematical simulation for the study of radionuclide kinetics in
the human body. Radiat. Prot. Dosim. 18 (4) (1987) 209.
Cristy, M., Eckerman, K.F.: Specific absorbed fractions of energy at various ages from internal
photon sources. ORNL/TM-8381/v1-7. Oak Ridge, TN: Oak Ridge National Laboratory, 1987.
ICRP: Radiation dose to patients from radiopharmaceuticals. ICRP Publication 53. Ann. ICRP 18
(1-4). Oxford: Pergamon Press, 1987.
Stieve, F.E.: Placental transfer of other nuclides. In: Gerber, G.B., Mtivier, H., Smith, H. (eds).
Age-related factors in radionuclide metabolism and dosimetry. Dordrecht: Martinus Nijhoff,
1987, p. 315.
Berkovski, V., Likhtarev, I., Ratia, G., Bonchuk, Y.: Internal dosimetry support system:
Multipurpose research computer code. Radiat. Prot. Dosim. 79 (1-4) (1988) 371.
ICRP: Individual monitoring for intakes of radionuclides by workers: Design and interpretation.
ICRP Publication 54. Ann. ICRP 19 (1-3) (1988).
ICRP: Limits for intakes of radionuclides by workers: An addendum. ICRP Publication 30, Part 4.
Ann. ICRP 19 (4). Oxford: Pergamon Press, 1988.
ICRP: Protection of the patient in nuclear medicine. ICRP Publication 52. Ann. ICRP 17 (4).
Oxford: Pergamon Press, 1988.
NEA/OECD: Gastrointestinal absorption of selected radionuclides. A report by an NEA expert
group. Paris: Nuclear Energy Agency/OECD, 1988.
Zankl, M., Veit, R., Williams, G., Schneider, K., Fendel, H., Petoussi, N., Drexler, G.: Radiat.
Environ. Biophys. 27 (1988) 153.
Birchall, A., James, A.C.: A microcomputer algorithm for solving first-order compartmental
models involving recycling. Health Phys. 56 (6) (1989) 857.
ICRP: Age-dependent doses to members of the public from intake of radionuclides. ICRP
Publication 56, Part 1. Ann. ICRP. 20 (2). Oxford: Pergamon Press, 1989.
Marshall, M., Stevens, D.C.: The purposes, methods and accuracy of sampling for airborne
particulate radioactive materials. Health Phys. 39 (1989) 409.
Tanaka, G., Nakahara, Y., Nakajima, Y.: Nippon Acta Radiol. 49 (1989) 344.
ICRP: 1990 Recommendations of the ICRP. ICRP Publication 60. Ann. ICRP 21 (1-3). Oxford:
Pergamon Press, 1991.
ICRP: Addendum 1 to Publication 53 Radiation dose to patients from radiopharmaceuticals.
ICRP Publication 62. Ann ICRP 22 (3). Oxford: Pergamon Press, 1991.
ICRP: Age-dependent doses to members of the public from intake of radionuclides: Part 2,
Ingestion dose coefficients. ICRP Publication 67. Ann. ICRP 23 (3/4). Oxford: Elsevier Science
Ltd, 1993.

Landolt-Brnstein
New Series VIII/4

7-70
93L1
94I1
94I2
94Z1
95D1
95H1
95I1
95I2
95S1

96D1
96E1
96I1
96I2
96I3
96P1
97A1
97I1
97I2
98I1
98N1
98P1
98S1
98U1
99C1

7 Internal dosimetry of radionuclides


Leggett, R.W., Eckerman, K.F., Williams, L.R.: An elementary method for implementing
complex biokinetic models. Health Phys. 64 (3) (1993) 260.
ICRP: Dose coefficients for intake of radionuclides by workers. ICRP Publication 68. Ann. ICRP
24 (4). Oxford: Elsevier Science Ltd, 1994.
ICRP: Human respiratory tract model for radiological protection. ICRP Publication 66. Ann.
ICRP 24 (1-3). Oxford: Pergamon Press, 1994.
Zubal, I.G., Harrell, C.R., Smith, E.O., Rattner, Z., Gindi, G., Hoffer, P.B.: Med. Phys. 21 (1994)
299.
Dorrian, M.D., Bailey, M.R.: Particle size distribution of radioactive aerosols measured in the
workplace. Radiat. Prot. Dosim. 60 (1995) 119.
Hubbell, J.H., Seltzer, S.M.: Tables of X-ray mass attenuation coefficients and mass energyabsorption coefficients, NISTIR 5632, Gaithersburg, MD: National Institute of Standards and
Technology, 1995.
ICRP: Age-dependent doses to members of the public from intake of radionuclides: Part 3:
Ingestion dose coefficients. ICRP Publication 69. Ann. ICRP 25 (1). Oxford: Elsevier Science
Ltd, 1995.
ICRP: Age-dependent doses to members of the public from intake of radionuclides: Part 4:
Inhalation dose coefficients. ICRP Publication 71. Ann. ICRP 25 (3-4). Oxford: Elsevier Science
Ltd, 1995.
Stabin, M.G., Watson, E.E., Cristy, M., Ryman, J.C., Eckerman, K.F., Davis, J.L., Marshall, D.,
Gehlen, M.K.: Mathematical models and specific absorbed fractions of photon energy in the
nonpregnant adult female and at the end of each trimester of pregnancy. Oak Ridge, TN: Oak
Ridge National Laboratory. ORNL/TM-12907, 1995.
Dimbylow, P.J.: Proc. Voxel phantom development 6-7 July 1996, Dimbylow, P.J. (ed.), Chilton,
UK: National Radiological Protection Board, 1996, p. 1.
EC. Council Directive 96/29EURATOM of 13 May 1996, laying down the basic safety standards
for the protection of the health of workers and the general public against the dangers arising from
ionising radiation. Off. J. Eur. Commun. 39, No. L159 (1996).
ICRP: Age-dependent doses to members of the public from intake of radionuclides: Part 5:
Compilation of ingestion and inhalation dose coefficients. ICRP Publication 72. Ann. ICRP 26
(1). Oxford: Elsevier Science Ltd, 1996.
IAEA: International basic safety standards for protection against ionising radiation and for the
safety of radioactive sources. Jointly sponsored by FAO, IAEA, ILO, NEA/OECD, PAHO and
WHO. Vienna, IAEA: Safety Series 115 (1996).
IAEA: Direct methods for measuring radionuclides in man. Vienna, IAEA: Safety Series 114 (1996).
Piechowski, J., Menoux, B.: Assessment of radioactive systemic uptakes by deconvolution of
individual monitoring results. Health Phys. 70 (1996) 537.
Ansoborlo, E., Boulard, D., Leguen, B.: Particle size distribution of Uranium aerosols measured
in the French nuclear fuel cycle. Radioprotection 32 (1997) 219.
ICRP: General principles for the radiation protection of workers. ICRP Publication 75. Ann.
ICRP 27 (1). Oxford: Pergamon Press, 1997.
ICRP: Individual monitoring for internal exposure of workers replacement of ICRP Publication
54. ICRP Publication 78. Ann. ICRP 27 (3/4). Oxford: Pergamon Press, 1997.
ICRP. Radiation Dose to Patients from Radiopharmaceuticals Addendum 2 to ICRP Publication
53, also includes Addendum 1 to ICRP Publication 72. ICRP Publication 80. Ann. of the ICRP.
28(3). Pergamon Press, Oxford (1998).
NRPB: Standards for intakes of radionuclides. Doc. NRPB 9 (4) (1998).
Phipps, A.W., Jarvis, N.S., Silk, T.J., Birchall, A.: Time-dependent functions to represent the
bioassay quantities given in ICRP Publication 78. NRPB-M824, Chilton, 1998.
Spitzer, V.M. and Whitlock, D.G: Atlas of the Visible Human Male. Sudburg, MA: Jones and
Bartlett (1998).
Ulanovsky, A.V., Eckerman, K.F.: Absorbed fractions for electron and photon emissions in the
developing thyroid: Fetus to five year old. Radiat. Prot. Dosim. 79 (1-4) (1998) 419.
Caon, M., Bibbo, G., Pattison, J.: Phys. Med. Biol. 44 (1999) 2213.
Landolt-Brnstein
New Series VIII/4

7 Internal dosimetry of radionuclides


99E1
99I1
99I2
99S1
99W1
00A1
00G1
00X1
01I1
01P1
01S1
01Z1
02I1
02I2
02I3
02I4
02N1
02P1
02S1
03C1
03E1
03G1
03H1
03K1
03M1
03P1
03S1

7-71

EPA.: Cancer risk coefficients for environmental exposure to radionuclides: Federal Guidance
Report No 13, EPA Number: 402R99001. Publisher Info National Service Center for
Environmental Publications, PO Box 42419 Cincinnati, OH 45242-2419, 1999.
ICRP: The ICRP database on dose coefficients: Workers and members of the public (CD-ROM).
Distributed by Elsevier Science Ltd, Oxford. ISBN 0-08-043876, 1999.
IAEA: Assessment of occupational exposure due to intakes of radionuclides. Safety Guide No.
RS-G-1.2, Vienna: IAEA, 1999.
Stather, J.W.: Dosimetry and effects of radioactive hot particles. Radiat. Res. Vol. 2: Dublin:
Proceedings of 11th ICRR, 1999.
WHO: Guidelines for iodine prophylaxis following nuclear accidents: Update 1999, Geneva:
WHO/SDE/PHE/99.6, 1999.
Anderson, E., Bai, Z., Bischof, C., Blackford, L.S., Demmel, J., Dongarra, J., Du Croz, J.,
Greenhaum, A., Hammarling, S., McKenney, A., Sorensen, D.: LAPACK Users Guide, SIAM,
Philadelphia, Third Edition (2000).
Gssner, W., Masse, R., Stather, J.W.: Cells at risk for dosimetric modelling relevant to bone
tumour induction. Radiat. Prot. Dosim. 92 (1-3) (2000) 209.
Xu, X.G, Chao, T.C., Bozkurt A.: Health Phys. 78 (2000) 476.
ICRP: Doses to the embryo and fetus from intakes of radionuclides by the mother. Corrected
version issued May 2002. ICRP Publication 88. Ann. ICRP 31 (1-3) (2001).
Polig, E.: Modeling the distribution and dosimetry of internal emitters: A review of mathematical
procedures using matrix methods. Health Phys. 81 (5) (2001) 492.
Saito, K., Wittmann, A., Koga, S., Ida Y., Kamei, K., Zankl, M.: Radiat. Environ. Biophys. 40
(2001) 69.
Zankl, M., Wittmann, A.: Radiat. Environ. Biophys 40 (2001) 153.
ICRP: Guide for the practical applications of the ICRP Human Respiratory Tract Model.
Supporting Guidance 3. Ann. ICRP 32 (1-2) (2002).
ICRP: The ICRP database of dose coefficients: Embryo and fetus (CD-ROM2). Distributed by
Elsevier Science Ltd, Oxford. ISBN 0-08-044188-2, 2002.
International Commission on Radiological Protection: ICRP Publication 89. Oxford, UK:
Pergamon Press, 2002.
Ishigure, N., Nakano, T., Enomoto, H., Matsumoto, M: Graphic Database on Predicted
Monitoring Data for Intakes of Radionuclide.
http://www.nirs.go.jp:8080/anzendb/RPD/gpmd.php (2002).
Nipper, J., Williams, J., Bolch, W.: Phys. Med. Biol. 47 (2002) 3143.
Petoussi-Henss, N., Zankl, M., Fill, U., Regulla, D.: Phys. Med. Biol. 47 (2002) 89.
Stather, J.W., Phipps, A.W., Harrison, J.D., Eckerman, K.F., Smith, T.J., Fell, T.P., Noke, D.:
Dose coefficients for the embryo and foetus following intakes of radionuclides by the mother. J.
Radiol. Prot. 22 (2002) 7.
Charles, M.W., Mill, A.J., Darley, P.J.: Carcinogenic risk of hot-particle exposures. J. Radiol.
Prot. 23 (2003) 5.
Eckerman, K.F., Ulanovsky, A.V., Kerr, G.D.: Electron and photon absorbed fractions in the
developing Fetus. ORNL/TM Report, 2003.
Guilmette, R.A., Durbin, P.W.: Scientific basis for the development of biokinetic models for
radionuclide-contaminated wounds. Radiat. Prot. Dosim. 105 (1-4) (2003) 213.
Harrison, J.D., Smith, T.J., Phipps, A.W.: Infant doses from the transfer of radionuclides in
mothers milk. Radiat. Prot. Dosim. 105 (1-4) (2003) 251.
Kramer, R., Vieira, J.W., Khouri, H.J., Lima, F.R.A., Fuelle, D.: Phys. Med. Biol. 48 (2003)
1239.
Mtivier, H.A.: New model for the human alimentary tract. Radiat. Prot. Dosim. 105 (1-4) (2003)
43.
Peace, M.S.: Practical experience of the application of ICRP models in internal dose assessment.
Radiat. Prot. Dosim. 105 (1-4) (2003) 33.
Stabin, M.G., Siegel, J.A.: Physical models and dose factors for use in internal dose assessment.
Health Phys. 85 (3) (2003).

Landolt-Brnstein
New Series VIII/4

7-72

7 Internal dosimetry of radionuclides

Acknowledgements
The authors are very grateful for the contributions from their colleagues in the development of this
review. They would particularly wish to thank Henri Mtivier, John D Harrison, Nina Petoussi-Hen and
Franois Paquet. They are also grateful for the excellent technical assistance from Karen Roberts in the
preparation of the manuscript.

Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

8 Decontamination

8-1

8 Decontamination

The first part of this Chapter describes decontamination techniques of large volume systems,
segmented parts and walls and floors. Advantages and disadvantages of the different methods are
discussed and examples of decontamination procedures and their results in laboratory- and large scale
are demonstrated. Considerations are focussed on nuclear facilities and power plants during operation
and decommissioning.
The second part deals with decontamination of the human skin. In case of contamination of the
human skin by radionuclides suitable measures have to be initiated to keep the dose to the skin below
the limits recommended by the ICRP. For purposes of dose estimates numerical values of the
equivalent dose rate in Sv/s at an activity per area of 1 Bq/cm2 are given for 128 radionuclides. In
addition first aid and specific decontamination procedures are described as simple decontamination
appliances immediately after contaminations or for decontamination of specific body regions and
organs below reference values for residual contamination.

8.1 Decontamination of materials


List of Abreviations
AGR
ALARA
AMDA
AP
APAC
APACE
BWR
CAN-DECON
CANDU
CEA
CEC
CEGB
CITROX
CORD
DF
EDTA
EPRI
Framatome
HX
LOMI

Landolt-Brnstein
New Series VIII/4

Advanced Gas Cooled Reactor


As Low As Reasonable Achievable
Automated Mobile Decontamination Application
Alkaline Permanganate
Alkaline Permanganate Ammonium Citrate
Alkaline Permanganate Ammonium Citrate EDTA
Boiling Water Reactor
CANdu DECONtamination
CANada Deuterium Uranium Pressurized Heavy Water Reactor
Commissariat l'Energie Atomic (France)
Community of European Countries
Central Electricity Generating Board
CITric Acid OXalic Acid
Cyclic Oxidation Reduction Decontamination
Decontamination Factor
Ethylene-Diamine-Tetraacetic Acid
Electric Power Research Institute (USA)
Company's Name
Heat Exchanger
Low Oxidation-state Metal Ions

8-2
MOPAC
NP
NPP
OZOX
PWR
SGHWR
WAGR

8 Decontamination

[Ref. p. 8-34

Modified Permanganat Ammonium Citrate


Nitric Acid Permanganate
Nuclear Power Plant
OZone OXalic Acid
Pressurized Water Reactor
Steam Generating Heavy Water Reactor
Windscale Advanced Gas Cooled Reactor

8.1.1 General approaches to decontamination


8.1.1.1 Contamination
Contamination is the deposition of the radioactive elements or compounds from a contaminant media or
gas, by chemical, physical or other ways, on the surfaces of components, systems, and structures in
nuclear facilities. The characteristics of the contamination are strongly correlated with the nature and
features of the surface and of the contaminant media. In metallic surfaces there often exists a chemical
similarity with the contaminant element (as for example for metallic cations in the water) that can cause
its diffusion into the metallic sub-layer, hence becoming very difficult to remove.
Contamination can be classified into three types:
- free contamination; i.e. can be removed by simple blowing, vacuum or similar methods;
- loose contamination; i.e. can be removed by common cleaning techniques;
- fixed contamination; i.e. cannot be removed without removal of surface layers.
The following presents typical contamination patterns encountered in nuclear facilities [88Int]. In reactor
systems, the radioactive contamination on the internal surfaces is caused by deposition from the reactor
coolant of neutron activated particles and dissolved elements, and of fission products and transuranics
released following a failure of the fuel cladding. These deposits become part of the oxide layer, which
forms on the inside of the piping. This layer has a complex structure, which depends on a variety of
parameters such as coolant chemistry, temperature of formation, system materials, operating time, etc.
Over long periods of time, the radionuclides in the layer can diffuse slightly into the base metal or
penetrate the pipes along grain boundaries. In general for water cooled reactors, two types of oxides form
on the internal surface of reactor piping: an adherent inner layer which is formed by in-situ corrosion of
the base metal, and a relatively loose outer layer which is formed by deposition or precipitation of crud
from the coolant.
8.1.1.2 Characteristics of oxide layer in BWRs and PWRs
Occupational dose in BWRs and PWRs is mostly caused by corrosion-originated nuclides: 60Co, 58Co,
54
Mn, 51Cr and 59Fe. Depending on fuel failure rate, the other fission species would contribute to plant
dose rates. Most part of these species is included in oxide layer inside pipes and equipment. Decontamination usually dissolves or removes the radioactive species together with the oxide layer.
Characteristics of the oxide layers are quite different between BWRs and PWRs. Iron occupies
80-90 % of metal elements in the BWRs oxide layer. BWRs use stainless steels and carbon steel for the
reactor cooling and the feedwater systems. PWRs use great amount of nickel-base alloys for steam
generators. Metal fraction of Ni and Cr is 60-70 % in PWRs oxide layer.
BWRs oxide layer grows in oxidising water chemistry and consists of -Fe2O3, Fe3O4, and NiFe2O4.
Reducing environment of PWRs forms (NixFe3-x-yCryO4)-type Cr-rich oxide layers. Table 8.1 compares
the oxide characteristics of Japanese BWRs and PWRs. Usually oxide forms indistinct double layer. The
inner layer grows from base metal and deposits tightly on the base metal surface. The outer layer contains
fuel surface crud released with shear stress by primary coolant flow. In some specific cases, the outer
layer crud is easily removed with ultrasonic vibration or high-pressure water jet.
Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

8 Decontamination

8-3

Table 8.1 Crud characteristics of Japanese BWRs and PWRs [00Hir1]


Primary coolant chemistry
Characteristics
of primary
system

Surface area percentage of


materials
Outer layer
Form

Crud analysis

Inner layer

Metal fraction

BWRs
Oxidising
SS:
38-42 %
Carbon steel:
16-20 %
Zircaloy:
40-44 %
Ni-base alloy:
<1 %
Fe3O4, NiFe2O4 (dominant)
Fe3O4 (dominant),
-Fe2O3, NiFe2O4,
FeCr2O4
Fe:
80-90 %
Ni:
7-10 %
Cr:
1-10 %

PWRs
Reducing
SS:4-6 %
Zircaloy:
25-28 %
Ni-base alloy
65-70 %
Other alloys
<1 %
Fe3O4 (dominant),
NiFe2O4, FeCr2O4
FeCr2O4,
Fe2CrO4
Fe:
Ni:
Cr:

20-40 %
25-60 %
15-45 %

8.1.1.3 Other types of contamination


In fuel reprocessing facilities, the acid process stream in the dissolution and separation steps of the
process, tends to inhibit the formation of an internal oxide layer and deposition of radionuclides is thus
limited. Nevertheless, after separation, the phase, which carries the uranium and plutonium, can form
pasty and heavy deposits in the pipes and tanks. These deposits are often very difficult to remove.
In other types of nuclear facilities such as hot cells and mixed oxide fuel fabrication plants, low levels
of contamination can exist in process vessels, cells, etc. as a result of normal operation. In UO2 fuel
fabrication plants low levels of activity are present from the processing of UO2.
300

Specific activity [Bq /g]

250

60

Co
137

Cs

200
150

Fig. 8.1 Penetration in concrete of 137Cs and 60Co in


samples of Gundremmingen KRB A Reactor (concrete
samples of floors were taken from the decommissioned
nuclear power plant). DS = decontamination seal; filled
columns and solid line: Cs-137; open columns and dasheddotted line Co-60.

100
50
0
DS 0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0
Penetration depth [mm ]

In most nuclear facilities, many external surfaces become contaminated as a result of leakage and
spillages from process systems and from demolition, maintenance and waste management activities.
Airborne or waterborne activity can deposit out forming contamination layers on floors, equipment,
instrumentation, etc. Thus surfaces can become contaminated by physical or chemical mechanisms.
Of particular concern is the potential contamination of concrete surfaces by waterborne
contamination. Unless the surface of the concrete is sealed, water-soluble radionuclides, such as 137Cs,
can penetrate deeply into concrete. The only method for removing such contamination would be to cut or
Landolt-Brnstein
New Series VIII/4

8-4

8 Decontamination

[Ref. p. 8-34

chip away the surface layer of concrete containing the radioactivity. To measure the penetration of the
contamination in Gundremmingen KRB-A, concrete surfaces were drilled and analysed by gamma
spectroscopy in the radiation protection laboratory to determine the nature and depth of the
contamination. 137Cs and 60Co, and traces of 134Cs, were found to be present. The isotope 137Cs accounted
for about 90 % of the radioactivity. As shown in Fig. 8.1, most of the activity is embedded in the
decontamination seal, but some contamination with a high proportion of 137Cs penetrates to a greater
depth than contamination in which 60Co is predominant [84Ebe].
For ventilation systems, the surface contamination is usually loose, although adherence is aided by oil
films often found on the inside of ducts particularly downstream of fans. Since the exhaust systems
operate at negative pressures they tend to draw in dusts and aerosols, which may contain activity.
Deposition tends to be heavier in sections of the ducting where the direction or velocity of the fluid
changes or at edges of joints or flanges.
For motors, instrumentation and walls etc., loose airborne contamination is usually the major problem.
This contamination can generally be removed if it is accessible. If motors and other delicate equipment
need to be reused, ultrasonic and freon decontamination processes are sometimes used.
8.1.1.4 Decontamination
Decontamination is the removal by chemical, physical or other methods, of surface radioactive material
from both internal and external surfaces of components, systems and structures in nuclear facilities.
Usually decontamination and cleaning are considered as separate processes even though they can often be
the same physical process; the difference is the degree of cleaning and the emphasis on species removed.
Decontamination is the removal of radioactive dirt and oxides from surfaces, whilst cleaning usually
refers to the removal of non-radioactive materials. Decontamination should be considered to be a part of
cleaning because, in general, only a small part of the material removed during decontamination is
radioactive. It is important to note that decontamination is not the elimination of the radioactivity, just the
removal to a different location. The term decontamination is widely used in reference to surfaces
commonly in contact with contaminated agents (such as reactor coolant, off gases, etc.) whereas the term
cleaning refers to surfaces only lightly contaminated by aerosols or by purge liquids, etc.
The concept of decontamination was introduced at the birth of the nuclear industry and was used to
describe the reduction of radiation levels on the surfaces of components, systems and structures in order
to allow their maintenance, repair, and control works. The importance of decontamination, and the
consequent development of new decontamination processes varied as the problems of reduction of
radiation levels and man-sievert expenditures (including costs) affected the exploitation of nuclear
stations and facilities.
In the early 1960s, decontamination was already a common practice in the nuclear industry. In the mid
1970s, with the support of regulatory agencies and industries, decontamination processes became more
sophisticated and a complete evaluation including environmental concerns, costs, legal and public
requirements became a common feature of decontamination practices. In the late 1970s, a new emphasis
was placed on the decommissioning of nuclear facilities and this introduced a new concept in
decontamination, not only to reduce radiation levels, which is normally the major objective of
decontamination, but also to facilitate waste management and, if possible, to permit reuse of the material
or components.
8.1.1.5 The use of decontamination in decommissioning
The techniques used in decontamination for decommissioning purposes have two main differences in
comparison with common in-service decontamination techniques. The techniques can be allowed to affect
the integrity of the base materials. This is because, in principle, the component or system will not be
reused. The techniques should generate the minimum quantity of secondary wastes.

Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

8 Decontamination

8-5

The basic approach to decommissioning work must be to answer the question: whether, when and how
decontamination is to be carried out. Hence, the decision to carry out decontamination is based on a compromise between the advantages and disadvantages of separating the radioactivity or dealing with the
fully active installation or component. Decontamination processes could be carried out merely to ease the
handling of materials or to bring the radioactivity down to a level where unrestricted release is possible.
The latter objective presupposes the existence of such a level, agreed upon with the relevant authorities.
Some examples are presented below of the above argument [90Ber]:
a)
b)
c)

A system or a component is to be worked on for inspection, maintenance or modification, and it


is so contaminated that the radiation levels in the work areas are above acceptable values with
reference to the established regulations or to the ALARA principle;
A facility, system or component is to be re-used for other purposes, which require it to be free
from contamination;
A facility, system or component is to be dismantled and the wastes arising from such proceedings are to be collected, conditioned, and disposed of in a safe and economical way according to
national practices and regulations, and consistent with an operational national waste management system.

In case (a), the objective is only to obtain a reduction in the level of the contamination without
damaging the components. Here the problem is to weight the overall decontamination costs, including
management of the necessary provisions for safe work in radiation areas, such as reduced individual
working hours, shields, remote operation, telemanipulation.
In case (b), complete decontamination is the main goal and the problem is a question of feasibility i.e. is it
possible to obtain the required level of decontamination, and of cost i.e. is it less expensive as a whole,
to decontaminate, or to dismantle everything and use new components?
Case (c) is common to all decommissioning strategies for nuclear facilities, although in some instances
cases such as (a) and (b) may arise. In this case, it may be necessary to consider in more detail, factors
that can influence the decision on whether to carry out decontamination in the first place. If
decontamination is the preferred option, then there is also a wide range of processes available to choose
from.
8.1.1.6 Identification of decontaminable components
From the above, it can be concluded that decontamination is a useful tool in decommissioning work but it
is not possible to generalise which parts should be decontaminated and, if so, how the decontamination
should be carried out. In reality a priority list of components and systems required for decontamination
needs to be drawn up [85Lr].
Components for decontamination should be identified as early as possible in order to avoid wasting
time and money on unnecessary decontamination work. Simple decontamination to remove weakly
adhesive contamination is still useful because it reduces radiation exposure and facilitates subsequent
handling.
One must first compare the two approaches, namely, decontamination for unrestricted release, or
direct transfer to some type of repository for radioactive materials. The exposure of personnel and the
respective costs of each approach must be considered. In addition to the main criteria above other factors
should be taken into consideration, these are:
The type and degree of contamination;
The geometry of the components;
The mass of the components to be decontaminated.

Landolt-Brnstein
New Series VIII/4

8-6

8 Decontamination

[Ref. p. 8-34

It is possible to determine the type and degree of contamination but careful consideration must be
given to whether components exposed for long periods to high pressures and temperatures (e.g. primary
circuit) can in fact be decontaminated, whereas components contaminated only by contact with air,
whether through moisture or other factors, clearly are decontaminable. In between lies a grey area, which
needs further investigation, based on operating data and/or tests, before a proper decision can be taken.
The geometry of the components plays a major role. Decontamination processes are mostly not able to
ensure even removal or cope with particular corners and cavities and these are the precise points at which
substantially higher contamination can occur. With complicated geometries, the measurements required to
produce evidence that the limits have been observed may not be possible or may only be carried out with
great difficulty.
The mass of components of the same type to be decontaminated, i.e. components that will be
subjected to the same decontamination process, is important in two ways. Firstly, there is little likelihood
that very large masses will be transported to a final repository, simply because they contain a few grams
or kilograms of radioactive material. Secondly, the decontamination process used must be suitable for
such large masses.
When selecting the process, one must ensure that the time needed for decontamination remains within
acceptable limits, since personnel costs, which are an important factor, increase in proportion to that time.
Where the amounts received from the sale of decontaminated material are significant the process need not
in principle spare the material; on the contrary it can be quite aggressive, since it is not planned to re-use
the material in its original form.
An estimate of the mass of material that might need to be decontaminated was made for two German nuclear power stations, one with a pressurised water reactor and one with a boiling water reactor. An effort
was made to make a distinction on the basis of the above-mentioned criteria. The material was divided
into three categories: nondecontaminable material, decontaminable material and material on which no
decision can yet be taken. Components, which today are still very difficult to decontaminate, or where
decontamination is not viable can naturally be reclassified at a later date on the basis of experience and in
the light of progress in decontamination techniques.
8.1.1.7 Effectiveness of decontamination, decontamination factor
The efficiency of different decontamination processes has to be evaluated. The common parameter is
called the decontamination factor. This is a numerical representation of the effectiveness of a decontamination process and it is calculated as ratio between predecontamination and post-decontamination
measurements, i.e.:
DF = Mb / Ma

(8.1.1)

Where DF = decontamination factor (generally greater than 1)


Mb = measurement before decontamination (at a reference point) and Ma = measurement after
decontamination (at the same reference point as Mb).
In terms of a percentage, the decontamination factor can be expressed as:
DF (%) =

Mb Ma
Mb

(8.1.2)

In the following, only the definition (8.1.1) will be considered.


With regard to the kind of measurements, which can be considered, the decontamination factor can be
defined (or calculated) by two different methods [85Duc].
The first method is to use radiation measurements. This is called the radiation DF and is defined as:
radiationDF =

Ib
Ia

(8.1.3)
Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

8 Decontamination

8-7

Where Ib = dose (radiation) rate before decontamination (at a reference point) and Ia = dose (radiation)
rate after decontamination (at the same reference point as Ib).
This definition is widely used in decontamination for operating plant, where the radiation
measurements referred to are dose rate area measurements; for this case the term man-sievert DF is also
used. In many cases the radiation measurements can be taken from monitor counters located near or over
the surface to be measured.
The second methods is to use activity measurements. This is called the decon DF and is defined as:
deconDF =

Ab
Aa

(8.1.4)

where Ab = activity before decontamination (at a reference point) and Aa = activity after
decontamination (at the same reference point as Ab)
This definition is widely used for off-line decontamination where the surface activity can be properly
measured. In laboratory studies and research, the decon DF is more widely used than the radiation DF.
As mentioned before, each numerical value of DF must be referred to a single measurement before
decontamination and a single measurement after decontamination. This means that the decontamination
factor can be of relevance only for single points or for very small surfaces or components (which can be
measured with a single operation). In any other case, where many measurements have been carried out it
is necessary to calculate an average DF.
The activity of the background is a parameter which is strongly dependent on the procedures and
instrumental techniques used for the activity measurements. For evaluation of Decontamination Factors
according to (8.1.3) shielding measurement devices are necessary to provide correct data.
8.1.1.8 Decontamination techniques (processes)
Decontamination techniques may be classified in several different ways depending on the purpose of
decontamination e.g. to save man-sieverts, for restoring the component/system, for decommissioning, the
kind of decontamination media e.g. chemical, mechanical, electrochemical, etc., and on the nature of the
surface required for decontamination (e.g. metal, concrete, painted surfaces, etc.).
The most widely used criterion refers to the kind of decontamination media. Nevertheless different
classifications have been proposed since in many cases it is not easy to clearly define the decontamination media. Some processes may combine several different decontamination media e.g.
electropolishing, which combines chemical and electrical actions, or water jets used with detergents,
which combine mechanical and chemical actions. In the following paragraphs some of the classifications
of decontamination techniques proposed by different studies are presented:
The US-Department of Energy (DOE) Decommissioning Handbook [80Man], in 1980, classified
decontamination techniques into four categories: (i) chemical decontamination, (ii) manual and
non-chemical decontamination, (iii) electropolishing, and (iv) ultrasonic/chemical decontamination
Chemical decontamination:
Alkaline Permanganate (AP), Ammonium Citrate (AC), EDTA, Oxalic Acid (OX), Citrox, Sulphamic
Acid, Hydrochloric Acid, Nitric Acid, Sulphuric Acid, Phosphoric Acid, Oxalic Peroxide (OP),
Sulphox, Can - Decon, NS - 1
High-pressure water lance
Electropolishing: in-tank and in-situ
Ultrasonic decontamination

Landolt-Brnstein
New Series VIII/4

8-8

8 Decontamination

[Ref. p. 8-34

Further Classifications were given by:


The Electric Power Research Institute (EPRI) [82Gar], in 1982,
The US-Nuclear Regulatory Commission (NRC) [81Nel], in 1981,
The Commissariat A l'Energie Atomique-France (CEA) [82Com] in 1982,
and the International Atomic Energy Agency (IAEA) [83Int], Vienna, in 1983.
8.1.1.9 Decontamination and secondary waste generation
The problem of secondary waste arising from decontamination works is of major concern in terms of
quantities and characteristics. This problem is of particular relevance for chemical decontamination
processes for decommissioning. In this context it became one of the leading factors for the real
acceptance of the process.
On-line chemical processes like Can-Decon, Cam-Derem, LOMI, CORD/OZOX and EMMA include
phases of solution cleaning in the process itself and generate relatively low volumes of ion-exchange
resins as secondary waste. With electropolishing decontamination using standard electropolishing
solutions, such as concentrated phosphoric acid, the treatment of the spent solution by reprocessing and
recycling the solutions in order to reduce the final volume of wastes for storage has to be considered.

8.1.2 Decontamination techniques for large volume closed systems


8.1.2.1 Reactor decontamination in BWRs and PWRs
8.1.2.1.1 Chemical decontamination principle
Chemical decontamination reagent work to dissolve spinel oxide involving Fe3+, Ni2+ and Cr3+ on the
inner surfaces of pipes and equipment. The three-valent ions have low solubility. The oxide can be
dissolved as follows:
Reducing dissolution: Fe-containing oxide is effectively dissolved with reducing reagent and acid
following the reducing dissolution scheme:
Fe2O3 + 6 H+ + 2 e 2 Fe2+ + 3 H2O
Fe3O4 + 8 H+ + 2 e 3 Fe2+ + 4 H2O
Acid dissolution: Acid can dissolve the spinel oxide. But acid alone also attacks the base metal:
Fe3O4 + 8 H+ Fe2+ + 2 Fe3+ + 4 H2O
Fe + 2H+ Fe2+ + H2
Oxidising dissolution: Cr-containing oxide is dissolved following the oxidising dissolution scheme with
permanganate ion:
Cr2O3 + 2 MnO4 + H2O 2 HCrO4 + 2 MnO2
A pH-potential diagram is illustrated in Fig. 8.2. Chemical decontamination is an oxide-dissolving
technique used not in the passive state area, but in the metal corrosive area (indicated with hatched line in
the Fig. 8.2).
Nearly all decontamination processes like Can-Decon, NS-1, LOMI and CORD reduce Fe3+ to Fe2+.
Fig. 8.1 shows the Redox-potential as a function of pH-value of nitric-acid-permanganate (NP) and
alkaline permanganate (AP). HP (permanganic acid) is in the same potential range of NP. All three
oxidation methods (HP, NP and AP) oxidize Cr3+ to Cr6+ but have no ability to dissolve Fe2O3 and Fe3O4.
NP solution is advantageous in the field of Fe/Cr/Ni - austenitic materials, AP in the field of Ni-alloys.
Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

2.0

8 Decontamination

H2CrO4

Cr2 O 27HCrO -4

1.6

Potential [ V vs.SHE ]

1.2

CrO 24

NP
b

8-9

log C = - 4
Cr :
Fe :
Ni :
Mn :

3+
2+
0.8 Fe / Fe

AP
NS -1

0.4
0

LND 101A
a

LOMI

Ni 2+/ Ni

Cr2 O3

- 0.4

Fe2 O3
Passive state

- 0.8

Fe 2+/Fe

Cr 3+/ Cr 2+

Fe3 O4

Cr 2+/ Cr

-1.2
-1.6
-2

Fig. 8.2 pH-potential diagram of chemical decontamination; [00Hir]

Cr (OH) 2

6
pH

10

12

14 16

Application of oxidation decontamination


Metal fraction of Cr in the oxide layer strictly affects decontamination performance. In the case of lower
chromium content than 7 %, reducing reagent achieves successful result. But in the cases of Cr content
higher than 7 %, very few reducing reagent works well without the steps of oxidising decontamination.
The oxidising reagent dissolves Cr and breaks spinel structure before the reducing step.
8.1.2.1.2 Testing material compatibility during and after decontamination
Before application of any decontamination process it has to be qualified in laboratory tests.
Table 8.2 lists evaluation issues to confirm materials compatibility during laboratory test. This basis
qualification covers the influence of the solvent during decontamination and the post operation behaviour
in the NPP systems. In addition during application on site test coupons can be inserted to the
decontamination circuit to monitor the corrosion and IGSCC. Occasionally, actual pipes are taken from
the plant and used for own compatibility evaluation.
Materials compatibility during decontamination
General corrosion is evaluated with weight loss measurement and surface/cut surface observation of test
coupons. General corrosion is a key issue of carbon steel and low alloy steels rather than stainless steels
and nickel base alloys. Flowrate of decontamination liquid, temperature and inhibitor effect should be
taken into account.
No galvanic corrosion effect is reported on the present existing decontamination methods for BWRs
and PWRs.
To evaluate pitting or inter-granular corrosion, new test coupons might be inappropriate. Probes from
actual tube materials that experienced plant operation history should be tested. Corrosion in crevices
caused by residual decontamination reagents should be evaluated.

Landolt-Brnstein
New Series VIII/4

8-10

8 Decontamination

[Ref. p. 8-34

Table 8.2 Material testing for decontamination reagent evaluation [00Kat]

During
decontamination

Evaluation issues
General corrosion
Pitting, crevice corrosion
Inter-granular corrosion
Long term compatibility against IGSCC
Crack growth rate of SCC

After decontamination

Corrosion with residual


decontamination reagent
Effect of decontamination repetition
Recontamination

Materials
Carbon steel
Carbon steel
Low alloy steel
Stainless steel
Ni base alloy
Stainless steel
Ni-base alloy
Stainless steel
Ni-base alloy
All reactor materials
All reactor materials
All reactor materials

Material behaviour after decontamination is one of the most concerning issues. Results of a systematic
survey conducted in western countries are described below.
United States
In 1980s, London Nuclear Ltd., GE and other organisations tested material compatibility to apply reactor
decontamination mainly for BWRs and Candu reactors. The tests were mainly on IGSCC susceptibility of
type 304 stainless steel (SS) and nickel base alloys (Inc 600). Can-Decon, PNS-Citrox, and LOMI were
tested as listed in Table 8.3 [00Kat1].
Can-Decon slightly increased SCC susceptibility of type 304 stainless steel, therefore the process was
modified to a process called CANDEREM.
8.1.2.1.3 LOMI, Can-Decon/CANDEREM and CORD / CORD UV
LOMI, Can-Decon/CANDEREM and CORD / CORD UV were the most important processes for the last
10 years, LOMI mainly in the US, CANDEREM in Canada and US, CORD UV in Western Europe and
Japan. In recent years in the US a revival of the CITROX process could be observed instead of applying
LOMI and CANDEREM.
LOMI was developed by CEGB with the target to dissolve only iron oxide (hematite) located on fuel
elements within the core of the SGHWR. By adding an AP and NP oxidation step the application range of
the process was extended mainly to BWR.
Until 1990s, Can-Decon was applied to BWR recirculation systems and other actual plants at least 34
times. After the evaluation regarding IGSCC, CANDEREM was developed. The CANDEREM uses
EDTA and citrate but no oxalate, which was the main chemical of Can-Decon. Both processes were developed by AECL. The CANDEREM is used at a temperature higher than 100 C.
In 1991, the CORD method developed by Siemens/KWU was applied to NPP Isar, a BWR in
Germany. The CORD process strictly controls pH-value and corrosion potential using oxalic acid as
reducing reagent. As a preoxidation step permanganic acid (HP) is applied. Many plants including
Japanese BWRs have applied CORD since 1991 to the present. Since 1994 the CORD process was
improved to the CORD UV process, in which the decontamination chemicals are destroyed during the
process to carbon dioxide and water. There has been no negative information regarding IGSCC with the
CORD/CORD UV process. Many plants have used this process. There is a lot of experience available on
this decontamination technique. Questions how circulate and to heat the solutions are well experienced.
As example Fig. 8.3 shows the Fukushima Daiichi, Unit 2 Flow Diagram for decontamination [00Wil]. In
total, more than 400 full system decontaminations have been performed with the Cord process.
Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

8 Decontamination

8-11

Table 8.3 Material-testing results for decontamination reagent evaluation in the US [00Kat1]
Decon reagent

Test
method
Tube test

304 SS

Can-Decon
(LND-101A)

SSRT

Can-Decon
(LND-104, LND101A)

Double Ubend
4-pointsupporting
bend test
SSRT

Can-Decon
(Nutek L-106,
LND-101)

Can-Decon
(LND-104)
Can-Decon,
LOMI

SSRT
Stress
beam Test

PNS-Citrox

SSRT

LOMI,
Can-Decon
PNS-Citrox

SSRT
Stress
beam Test

LOMI

Tube test
Crack
growth rate
measurement
SSRT

Can-Decon
(LND-101A)

Landolt-Brnstein
New Series VIII/4

Materials

Water
chemistry
BWR
(DO: 8ppm)

Results

304 SS

BWR
(DO:
0.2ppm)

304SS,
Inconel
600

PWR
(DH:
>3ppm)

No effect on SCC
susceptibility in 100 h
decon, but 500 h decon. Increased on sensitised 304 SS.
No susceptibility increase to SCC

304SS,
Inconel
600
304SS,
Low alloy
steel,
(SA533B)
Inconel
600

PWR
(DH:
>3ppm)
BWR
(DO:
0.2ppm)

No susceptibility increase to SCC

304SS,
Low alloy
steel,
(SA533B)
Inconel
600
304SS,
Inconel
600

BWR
(DO:
0.2ppm)

Sensitized 304SS increased susceptibility


to SCC in some cases.

BWR
(DO:
0.2ppm)

Can-Decon and PNSCitrox increased susceptibility of sensitised


304SS to SCC

304 SS
316 NG
SS

BWR
(DO:
0.2ppm)

No susceptibility increase to SCC

304SS

BWR
(DO:
0.2ppm)

No susceptibility increase to SCC

No acceleration on
propagation rate of
existing crack

Can-Decon increased
304SS SCC susceptibility. No effect with
LOMI

Corrosion

Reporting
organisation
London
50m IGA
was observed Nuclear Ltd.
on sensitised [85EPR]
304 SS.
No descrip- Ontario
tion
Hydro
[85EPR1]
Pitting on
304 SS and
1.5m IGA
symptom on
Inconel 600
No IGA

London
Nuclear Ltd.
[85EPR2]
London
Nuclear Ltd.
[85EPR3]
G.E.
[86EPR]

Can-Decon
caused
200m IGA
on 304SS and
100m IGA
on Inconel
600. No IGA
with LOMI.
G.E.
80m IGA
on 304SS and [86EPR1]
60m IGA
on Inconel
600
IGA were
observed as
the same
extent as the
above two
columns
No description

G.E.
[86Man]

G.E.
[86Man]

Slight IGA
G.E.
symptom was [86Man]
detected in
some cases

8-12

8 Decontamination

[Ref. p. 8-34
Existing Recirculation
System

Vent
UV Modules

Temporary Decon.
Equipment

Spray
Ring

Chemical
Injection
Tank
Chemical
Injection
Tank

RPV
Pump
20% speed
Reactor
Recirculation
System
Pump

CRD Housing
Decon Loops:
200 m3/ h /each

Demineralizer

Main Circulation
Pump

Heater

Cooler

Fig. 8.3 Fukushima 1 Unit 2 Flow diagram; [00Wil]

8.1.2.2 Fuel assemblies and decontamination


Fuel surface crud usually contains a 100 to 1000 times higher inventory of radioactivity than recirculation
system oxide layers. A decontamination reagent is easily decomposed by gamma rays and neutrons from
the fuel even in reactor lay down periods. In spite of these difficult conditions fuel elements may have to
be decontaminated for two reasons. In case the crud deposits cause too high pressure drops to achieve a
homogeneous reactor coolant water flow cleaning is required. In addition, the treatment of spent fuel elements depends on a certain degree of cleanliness.
The SGHWR (100 MWe, closed in 1991, UK) and Candu reactors often performed the
decontamination of fuel assemblies in situ. The LOMI developed for this purpose has self-regenerating
ability in the presence of radiolysis. Pressure-tube-type reactors are usually designed for on-power
refueling. This requires more time than required for BWRs and PWRs to discharge all fuel from the core.
In spite of that, Fugen (165 MW, Japan) a pressure tube type reactor, was decontaminated after
discharging the fuel to obtain a higher decontamination factor and to reduce radioactive waste.
NPP Paks found a thermal-hydraulic anomaly in the reactor core caused by corrosion product deposits.
Consequently, the coolant flow through the fuel assemblies was insufficient resulting in a temperature
asymmetry in the reactor core. The fuel assemblies were removed from the core and successfully cleaned
applying the CORD UV process.
8.1.2.3 Decontamination of sodium cooled systems
These systems may be decontaminated effectively by acid solutions. Within the research and
development programme of the CEC an inorganic acid-based process has been evaluated and tested by
CEA for the RAPSODIE reactor in France [89Cos]. Decommissioning operations began in 1987.
Preliminary cleaning and water rinsing after isolation of the main vessel eliminated all traces of residual
sodium. Main contamination nuclides were 137Cs, 63Ni and 54Mn.
After steam-cleaning to remove the residual sodium, the specimens were highly rusted. One of the
first reagents to provide satisfactory results was a mixture of nitric acid and sulphuric acid at 85 C. In
order to improve the effectiveness of the decontamination, the aggressiveness of the reagent was
Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

8 Decontamination

8-13

enhanced by adding cerium (IV) in sulphate form. This reagent is sufficiently oxidising (Redox
potential Eh = 1.610 V) iron, chromium and nickel in austenitic steels. Together with an alkaline
washing excellent decontamination results were obtained: The estimated initial contamination level of
5500 Bq cm2 was reduced to less than 10 Bq cm2. The low residual contamination values allowed to
estimate the pipes to be melted down for reutilization and release and an authorisation has been
applied for. The dose rates were uniform throughout the facility, ranging from 1 to 15 Gyh1
8.1.2.3 Gas cooled reactors (WAGR)
An important feature of the Windscale AGR dismantling programme was the removal and disposal of the
four heat exchangers [89Cro]. Each heat exchanger contains tube banks (or superheater banks), with plain
Cr/Mo low alloy steel tubes and evaporator and economiser banks with finned mild steel tubes.
Contamination was found to be incorporated in this oxide layer and to consist predominantly of 137Cs
with some 134Cs and 60Co. Average values of 1.6 103 Bq cm2 for Cs and 0.2 103 Bq cm2 for Co were
measured, giving a total for the superheater of about 2 1011 Bq. To remove the activity it was necessary
to remove the oxide layer from the tubes.
The decontamination was performed by spraying with a 3000-litre mixture of 0.5 molar (3.15 %
by weight) nitric acid and 0.0025 molar citric acid at ambient temperature. In total the radiation level
was removed by 70 %, activity were removed, the manSv-uptake was reduced remarkably and the
targets reached.

8.1.3 Decontamination techniques for segmented parts


8.1.3.1 Chemical decontamination
The chemical decontamination of an item removed from a nuclear plant or facility is generally carried out
by immersion in a tank containing the chemical reagent [92Com]. The size of the tank depends on the
dimensions of the item to be decontaminated. A common size is one, which is 1-2 m square with a depth
of 0.5-1 m. Tanks for water rinsing are always installed. In sequential multistep processes the availability
of several tanks can be useful in order to reduce the time needed.
Chemical decontamination is characterised by the following parameters:
Type and nature of the chemical reagent;
Temperature of the process;
Duration of the process.
The effectiveness of decontamination can be improved by increasing the duration of the treatment and
the temperature. Optimum results are usually obtained with the solvents at elevated temperatures (up to
120 C). During the decontamination process, as the concentration of the contaminants in the solution increases, the item being cleaned may become re-contaminated. This problem can be minimised by
cleaning the least contaminated items first and by cleaning or replacing the solution if the concentration
of contaminants exceeds certain levels.
It should be noted that strong corrosive attack of the base metal may not result in high
decontamination factors. These can however be achieved without significant corrosion of the base metal.
Strong solutions of nitric and phosphoric acid used in the USA Bonus programme resulted in the removal
of up to 0.2 mm of the inner wall of pipes, but only an average decontamination factor of approximately
100 was achieved. Results to date with non-aggressive processes indicate that decontamination factors as
high as 2000 can be achieved without significant corrosion of the base metal [81Nel].

Landolt-Brnstein
New Series VIII/4

8-14

8 Decontamination

[Ref. p. 8-34

Some multistep processes are commonly used for removing highly adhesive contamination layers. In
many cases chemical decontamination can be used as a single step in complex processes e.g., before electropolishing, items covered with thick oxide layers are submitted to chemical decontamination in order to
soften the oxide.
8.1.3.1.1 Chemical reagents
Since the 1950s several chemical reagents have been commonly used for cleaning contaminated items.
Lists with more than 100 chemical products can be read in dedicated decontamination handbooks [88Int,
81Nel, 82Com]. Some examples of chemical reagents used are given in Table 8.4.
Table 8.4 Some reagents typically used in chemical decontamination of reactor components depending
on type of material to be decontaminated.
Materials
Reagents
Aluminium
HNO3, Na-EDTA + 2 % detergent, 10 % citric acid, sulphamic acid .
Brass
HNO3, 5 % AC
Carbon steel
Inhibited HCl, inhibited sulphamic acid, EDTA, citric + oxalic acids, APAC,
CITROX,
Copper
Phosphorous, nitric and acetic acids
Nickel & Alloys
AP, 25 % HNO3 + 25 % HF, AP + AC, AP + CITROX, EDTA
Monel
25 % sulphamic acid
Stainless steel
AP- CITROX, EDTA-CITROX, 30 % HF + 20 % HNO3, 70-80 % H3PO4,
0.4 M Cr SO4 + 0.5 M H2SO4, AP-HNO3, AP-AC, AP-OX, APACE
Zircalloy
OX + H2O2 + glucosic acid, EDTA, 8 M HNO3,
AP: alkaline permanganate, AC: ammonium citrate, OX: oxalic acid, CITROX: citric + oxalic acid,
APACE: AP + AC + EDTA.
There is a wider range of solvents to choose from for decommissioning programmes since corrosion
of the base metal is of little concern. Certain solvents exhibit a time dependency in the mixing, heating,
recirculation and draining cycle that affects both the chemical solution stability and the solubility of contained contamination. Each process under consideration would have to be evaluated for the effect of a
loss-of-flow accident and associated cooling of the solvent. Factors considered would include toxic or
explosive gas generation, excessive plate-out and excessive corrosion. The selected process must include
appropriate emergency procedures, e.g. emergency draining, gas detection, and emergency ventilation.
8.1.3.1.2 Spent decontaminant solutions
The selection of the chemical reagent directly affects the features of the secondary wastes arising
from the process. It is obvious that continuously renewing the solution increases the decontamination
effectiveness [85Pas] but the quantity of spent solution to treat and to dispose of also increase
dramatically. In latter years the regeneration of chemicals have become a fundamental step in all
chemical decontamination processes.
Several conventional chemical processes can be used for regenerating the spent solutions either on
their own or in combination and they include: ion exchange, evaporation/distillation or electrodialysis.
The problem of limiting the secondary wastes arising from the decontamination process, sometimes
leads to the selection of other similar processes like electropolishing or ultrasound with chemicals rather
than chemical decontamination. As stated previously, only a detailed cost/benefit analysis can provide the
actual criteria for selecting the best option for decontamination.
Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

8 Decontamination

8-15

8.1.3.2 Electrochemical decontamination


8.1.3.2.1 In-Tank
Electrochemical decontamination can be considered in principle to be a chemical decontamination
assisted by an electrical field. Nevertheless the best electropolishing is a process widely used in nonnuclear industrial applications to produce a smooth polished surface on a variety of metals and alloys. It
can be considered the opposite of electroplating as metal layers are removed from a surface rather than
added as a coating. Usually the object to be electropolished is immersed in a tank of electrolyte and is
used as the anode in the electrolytic cell. The passage of electric current results in the anodic dissolution
of surface material and, for normal operating conditions, a progressive smoothing of the surface.
A progressive dissolution of the surface material occurs within a certain range of voltage and current
density [83Int]. If the voltage and current densities are too low, the surface is attacked non-uniformly,
causing etching rather than polishing. Similarly voltages that are too high cause severe pitting of the
surface [87Pav].

(+)

DC
Power
supply

( -)

Anode

Cathode
Hydrogen
Oxygen

Containment
Tank

Electrolyte

aa
aaaa
aa

aa
a
Fig. 8.4 Electropolishing device
Surface to be
decontaminated

If the anode is a contaminated material such as metal or alloy, all radioactive contamination on the
surface (or entrapped within surface imperfections) can be removed and released into the electrolyte by
this surface dissolution process [85Pas]. The process produces a very smooth (0.02-0.03 mm),
non-reactive and non-adsorbing surface resistant to recontamination during further operations. Experience
has shown that electropolishing is an effective technique for removing both fixed and smearable
radionuclide contamination. Moreover it is fast and easily controlled.
In general, there are two methods of application for electropolishing. The most common method is
immersing the item to be decontaminated in a tank filled with a suitable electrolyte. The second method
involves the use of in-situ mobile devices that are able to electropolish part of the surface of the item,
which, because of size or installation, cannot be electropolished in a tank.
Phosphoric acid is normally used as the electrolyte in electropolishing because of its stability, safety
and applicability to a variety of alloy systems. Moreover, the non drying nature of phosphoric acid helps
minimise airborne contamination, and the good complexing characteristics of phosphoric acid for metal
ions is a significant factor in minimising recontamination from the electrolyte.
Representative operating conditions for decontamination using phosphoric acid electrolytes are:
solution temperatures of 5 to 25 C, phosphoric acid concentrations of 40 to 85 %, electrode potentials of
8 to 12 V and current densities of 5 to 25 A cm2.
Landolt-Brnstein
New Series VIII/4

8-16

8 Decontamination

[Ref. p. 8-34

The direct current power supply converts alternating current in to direct current which generates the
current flow between anode and cathode required for the electrochemical reaction. Voltage requirements
range from 0 to 24 V with sufficient amperage to provide the required current densities.
From experience gained in non-nuclear industrial applications, electropolishing normally uses
phosphoric acid as the electrolyte (sulphuric acid is an alternative). However, during the last decade a
variety of different electrolytes have been tested and developed with particular reference to the problem
of reducing the secondary wastes arising from the process. As a consequence new processes usually
called electrodecontamination or electropickling decontamination have been proposed. These consider the
use of basic solutions as well as organic acid mixtures. Finally, among the electrodecontamination
processes for in situ applications, electrobrushing using an electrobrush continuously fed by an
electrolyte should be mentioned.
Electrolytic decontamination can be used to remove fixed or imbedded contamination on iron-based
alloys, including stainless steel, as well as on copper, aluminium, lead, and molybdenum. However the
effectiveness of the decontamination can be limited by the presence of foreign materials on the surface of
the items to be decontaminated. Materials such as oil, grease, oxide (rust), and paint or other coatings
should be removed before decontamination.
In decontaminating (mainly for decommissioning purposes) reactor coolant systems, the systems are
usually covered by oxide layers that in principle work as a barrier for electropolishing. This problem can
be overcome by increasing the electropolishing time. Nevertheless some new processes consider the
periodic switching of polarity between cathode and anode, as well as changing voltage and current in
order to increase the removal of the surface materials [86Gau]. Generally, at least two (stainless steel)
tanks are required for performing electropolishing. One tank contains the electrolyte, electrodes, and parts
to be decontaminated (as anode). The other tank holds the water used for rinsing the parts after
decontamination. Power supply amperage capacities up to 2700 A are common.
The cathode is normally a piece of copper, or stainless steel, positioned in the electrolyte within
30-100 mm from the item to be decontaminated. In addition for special items, the walls of the tank for
immersion electropolishing can also serve as the cathode.
To control vapours released from the electrolyte during the electropolishing process an extraction
hood is located alongside the electropolishing tank. Provision for heating and agitating the electrolyte and
rinse tank is also required.
Studies on in-tank electropolishing became of relevance in the early 1970s in the USA where they
were used to decontaminate hot-cells, glove-boxes, and tools contaminated by alpha emitters.
Decontamination carried out in conjunction with Rockwell Hanford Operations and United Nuclear
Industries in the USA, show that components heavily contaminated with PuO were decontaminated from
1 million dpm per 100 cm2 to background in less than 10 minutes [78All].
Typical decontamination times range from 5 to 30 min, corresponding to the removal of 10 to
50 mm of surface material at a current density of 2-15 A/dm2. It is usually necessary to remove the anode
contacts at least once during a cycle in order to decontaminate the area under the contacts.
Since the early 1980s commercial use of electropolishing in decontamination of reactor coolant
water systems and components for decommissioning purposes was made at the KRB power station
(reactor A) in Germany [83Eic, 01Eic].
Electrochemical decontamination by electropolishing causes a steady increase of dissolved iron in the
phosphoric acid. If the content of iron exceeds 100 g dm3, a precipitation of iron phosphate occurs and
this stops the efficiency of the decontamination process. Therefore the acid has to be exchanged or
regenerated periodically. The regeneration of phosphoric acid is based upon the reaction of Fe2+ with
oxalic acid (see Fig. 8.5). Electrochemical decontamination of steel, however, generates a high percentage
of Fe in the phosphoric acid, which cannot be precipitated to iron oxalate. The high content of Fe3+ is
reduced to Fe2+ by subsequent pickling. When a high portion of Fe2+ is obtained, the phosphoric acid has
to be mixed with an aqueous solution of oxalic acid. The activity (mainly 60Co) is mostly separated from
the solution by precipitation together with the iron. The iron oxalate is dried and stored for subsequent
processing. The initial concentration of the phosphoric acid can be achieved by an evaporation process
[89Sta].

Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

8 Decontamination
Oxalic Acid

Decontamination of
1 Mg Steel Material
+
-

8-17

Iron Oxalate

Precipitation
Thermolysis

H 3 PO 4
Chemical,
Electrochemical

Vaporization

Waste:
15 kg Iron Oxide

Fig. 8.5 Flow chart of the


phosphoric acid regeneration

Several electrolytes were investigated and proposed as alternatives to phosphoric and sulphuric
acid. The need for new electrolytes was initially motivated by the incompatibility of phosphoric and
sulphuric acids with the existing treatment facilities and by the possibility of generating secondary
liquid wastes which are more easier to process.
This regeneration process was extensively proved at KRB Gundremmingen in Germany where more
than 200 m3 of phosphoric acid (concentration 20-40 %) have been regenerated. The iron oxalate can be
converted to iron oxide by pyrolytic decomposition.
With this method, KRB-A-Reactor materials like pipes, pumps, and housings were decontaminated.
Decontamination Factors of more than 100000 were reached.
8.1.3.2.2 In-situ
In the case of in-situ electrochemical decontamination, the surface of the item to be decontaminated is
flooded with electrolyte through a gap between the cathode of the device and the item's surface. The
inside of tanks, housings and other relatively open vessels, without internal components, can be
decontaminated without removal using an expandable bladder with a conductive surface that serves
both as the cathode and as a displacement device to minimise the electrolyte volume. Several devices
have been developed in the USA for application of in-situ electropolishing [82Gar1]:
The internal cathode device consists of a perforated, tubular, copper or stainless steel cathode section
with insulator-spacers at each end and has the provision for pumping the electrolyte and feeding power
through the insulator at one end. The perforated tubular section permits flow of the electrolyte to the pipe
surface being decontaminated, thus accomplishing the electropolishing action. An improved device with
four module heads was designed and used in Germany for the Obrigheim power plant [84KWU]. The
pump stream device consists of a perforated, disc-shaped, copper or stainless steel cathode facing the
surface to be decontaminated, with an insulated handle for flow of electrolyte and supply of power. The
electrolyte flows out of the end of the device in a stream and impinges on the surface being
decontaminated.
8.1.3.2.3 Electrobrushing
Electrobrushing is an electrodecontamination process for selected areas. The item to be decontaminated is
used as the anode, while an electrobrush serves as the cathode. The brush itself is a cellulose sponge
wetted by a continuous feed of an electrolyte, such as 5 % sulphuric acid solution inhibited with 1 g dm3
ethyl quinolinlium. Decontamination is carried out by scrubbing at a current of 15 to 40 A at 15 to 20 V,
and decontamination factors of around 30 are reported at a rate of 0.6 m2h1 [81Nel].

Landolt-Brnstein
New Series VIII/4

8-18

8 Decontamination

[Ref. p. 8-34

The disadvantages of this process include the production of large volumes of aqueous radioactive
waste and excessive attack of the surface by the electrolyte. In addition, if the decontamination is
performed manually rather than remotely, the radiation exposure to operators may be high.
Single electropolishing and brushing processes can be combined. Subsequently, in decontaminating cast steel components at the KRB Gundremmingen power station in Germany, it was found
that brushing prior to electropolishing resulted in a 35 % reduction of the time required for the
galvanic decontamination process and the reduction of dissolved iron in the electrolyte was also found
to be a benefit of prebrushing [89Sta].
8.1.3.3 Jetting decontamination techniques
The impingement of either a liquid or a solid media (or a liquid solid mixture called slurry) can be
successfully used for decontamination. Extensive use of jetting methods to clean surfaces and items has
been made in many industries and for varied applications. As a result of this several decontamination
methods have been studied, developed and as a result of this several contaminated materials.
Generally, jetting decontamination processes have a high flexibility and can therefore be applied to
both large surfaces e.g. floors and walls, and relatively small-contaminated items and systems.
Contaminated glove-box internals and several types of tools have also been cleaned using jetting
processes.
The problem of amount and characteristic of secondary wastes is one of the main concerns for jetting
processes. The amount of waste can be strongly reduced by recirculating, treating and rinsing the
impinging jetting media. Particular care must be applied to processes using abrasives.
A variety of nozzles and lance configurations can be used for high-pressure water cleaning depending
on the configuration of the item to be decontaminated. A straight jet can be used on the end of a long
handled lance to reduce worker exposures for decontamination of accessible tank interiors, walls or
floors. Self-propelling mole nozzles on a flexible high-pressure lance or hose can be used to
decontaminate the inside of tubing or pipes.
Because high pressure water cleaning is very effective for the removal of smearable surface
contamination, water lances have been successfully used to decontaminate pump internals, valves, cavity
walls, spent fuel pool racks, reactor vessel walls and heads, fuel handling equipment, feedwater spargers,
floor drains, sumps, interior surfaces of pipes and storage tanks.
Although decontamination factors of up to several hundred are commonly achieved, the normal factor
for most applications is up to 50. Decontamination factors of 2 to 50 with water as the agent and of 40
with the addition of a proper cleaning agent were achieved at a particular site [79Rem].
8.1.3.3.1 Abrasive jetting
Abrasive jetting is a very effective decontamination method in which an abrasive medium is propelled by
a jet of air (dry blasting) or water (wet blasting) against the surface to be cleaned [82Gar, 81Rem].
Typical abrasives are sand, alumina and metals, metal oxides and sawdust. Sand is the most common
abrasive because it is inexpensive and a good scouring agent.
Abrasive cleaning can be wet or dry. Abrasive particles are impelled at high velocity against the
surface to be cleaned by air, water or a mixture of the two, as in the following air abrasive blasting, water
abrasive blasting, air slurry blasting. Alternatively the particles may be carried in a viscous matrix and
rubbed against the surface (liquid honing or abrasive slurry cleaning) or the abrasive may be in the form
of stones, which can be used to grind or hone the surface to be cleaned. For effective cleaning either highpressure air at about 1 MPa or water at pressures similar to those used in hydrolaser systems are used,
depending on the application. The abrasive can also be attached to a flexible backing to form a type of
sandpaper or emery cloth or it can be forced against the surface by centrifugal action.

Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

8 Decontamination

8-19

The different types of abrasive can be grouped into three general classifications depending on
hardness:
Hard abrasive materials: For decontamination, hard abrasive materials (harder than the material to be
removed) are commonly used [64Ame]. In addition, there are three special sub-classifications:
Cleanable abrasives, soluble abrasives and system compatible abrasives. The last one being important
only for the decontamination of items required for reuse.
A cleanable abrasive simplifies the problem of secondary wastes by markedly reducing the quantity of
contaminated material to be disposed of after decontamination. One type of cleanable abrasive that can be
used on non-stainless steel surfaces is steel shot. To facilitate cleaning the particles should be smooth;
unfortunately smooth particles also reduce the abrasive action.
A soluble abrasive is a material, which can be used in solid form in an air or water jet and can later be
removed from the system by dissolving it in a liquid and flushing it out of the system. Boron oxide, which
dissolves in water to form boric acid, is such a material.
There are a number of methods of application of abrasive jetting, which depend on the carrier fluid,
flow velocity and kind of abrasive used. Abrasive blasting is often carried out in special cabinets [91Bru].
A specific case of abrasive blasting is dry ice blasting which uses ice pellets (2-3 mm in diameter)
produced by CO2 flashing at 40 C. The use of dry ice pellets as an abrasive media strongly reduces the
volume of secondary wastes produced [70Ayr].
The vacuum blasting method is a modification of air abrasive blasting for in-situ use where the
discharge nozzle is surrounded by a concentric hood. An air exhaust line is attached to the hood and the
blast air, debris and spent abrasive are drawn out through the exhaust line. The debris and abrasive are
separated and the abrasive is reused. Smaller, hand-held units are also available.
Water abrasive blasting has been used successfully to decontaminate a wide variety of contaminated
components. Methods of remote application have been developed for in-situ cleaning to supplement the
more common manual application methods. Decontamination factors of 200 to 300 are commonly
achieved.
8.1.3.3.2 Freon jetting
Systems have been developed to remove loose contamination from surfaces and equipment using
commercial freon (trichlorotrifluoroethane) cleaning solvents [88Int]. Freon has a low viscosity and
surface tension, which allows it to penetrate into cracks and crevices and remove contamination,
including that associated with grease, oil, etc. Freon is non-flammable and chemically inert and can
therefore be used to clean many types, of equipment without damaging delicate components. Most
radioactive contaminants are insoluble in freon and can be removed by filtration or distillation, allowing
recycling of the freon. Freon decontamination is carried out by directing a high pressure (15 MPa) jet of
the liquid onto the surface to be cleaned. The decontamination is usually carried out inside a glove box
[81McV], but experimental units have been developed for in-situ cleaning.
The freon liquid and particles of contaminant are collected in a sump; the liquid is then filtered to
remove the contaminants, cleaned and recycled. The freon is distilled as required to remove any
radioactive material, which has dissolved in the liquid. The use of this decontamination technique is often
limited due to legislative and regulatory restrictions in the industrial use of freon and freon-compounds
due to their potential effect on the environment.
8.1.3.4 Ultrasonic decontamination
Ultrasound consists of longitudinal mechanical waves and has been used over a long period of time for
cleaning dirty surfaces in non-nuclear industry. In particular, ultrasonic cleaning has been used with good
success for removing oil, grease, dirt and scale from a variety of items of various sizes and
configurations. As a result of this ultrasound was used as one of the first methods for surface
decontamination purposes in the nuclear industry [81McV].
Landolt-Brnstein
New Series VIII/4

8-20

8 Decontamination

[Ref. p. 8-34

The process is particularly appropriate for decontaminating items with complex surfaces where other
decontamination techniques are not suitable. It has been successfully used to clean dirt from holes, cracks
and crevices in parts made of metal, glass, and a variety of plastics. Its most common application is for
decontamination of tools and items that are evenly contaminated and it is carried out by immersion in an
ultrasonic tank containing water (or water with chemical additives) [82Gar]. New applications have been
developed replacing water with more aggressive chemicals in order to combine the cleaning effect of both
the chemical and ultrasound.
8.1.3.4.1 Ultrasonic technique
The two main components of ultrasonic cleaning systems are the ultrasonic generator and the transducer
(or vibrator). The ultrasonic generator converts normal 50-60 Hz power to a high frequency supply
usually in the range of 18 to 25 kHz. The high frequency alternating current is then fed to a transducer to
produce vibration in the liquid. The transducer is generally made of piezoelectric material (i.e. material
that will elongate or contract depending upon the polarity). As a consequence of the vibration,
compressive and rarefacted phases are present in the solution. In the rarefaction phase, cavities are
generated (mainly originating from nucleation points) and during the compressive phase these collapse
causing a phenomenon called cavitation. When an item is immersed in the solution, the collapse of
cavities causes scrubbing on its surface and hence produces a cleaning action. It is important to note that
the presence of many nucleation points causes cavitation mainly on the surface of the items. The
cavitation occurs even if the surfaces are inhomogeneous and complicated or located in inaccessible
zones. Calculations indicate that during cavitation localised peak pressures as high as 70 MPa can be
reached. These conditions produce a strong cleaning action on any surface upon which they act [82Gar].
Two factors play a fundamental role in the action of ultrasound: the cavitation threshold and the
scrubbing factor. The cavitation threshold is the pressure difference inside the fluid, which allows the
cavitation phenomenon to take place, and is directly correlated with the ultrasonic power applied to the
solution.
8.1.3.4.2 Ultrasound in conjunction with chemicals
It is well known that in order to increase the effectiveness of the process in terms of scrubbing effects, or
decontamination factors, an appropriate liquid should be selected. In ultrasonic cleaning, specific
chemical agents are commonly added and the ultrasonic cleaning combines the effect of cavitation of a
liquid at the surface to be cleaned with the chemical action of the liquid.
The physical and chemical properties of the liquid are important. Decontamination factors as low as 2
have been obtained by using pure water at room temperature. The addition of a cleaning or wetting agent
in the amount of 2 to 5 % by weight, with an increase in temperature to about 80 C, can greatly increase
the cleaning ability of water. The addition of small amounts of citric acid, or other chemicals, can also
enhance the cleaning ability.
Reports on the effectiveness of ultrasonic decontamination from the 40 or more nuclear plants where
it has been used are mixed. Some plants use it on a regular basis with good success, obtaining
decontamination factors in the range of 5 to 100 [80Man]. Other plants report little success, and some
have stopped using ultrasonic cleaning entirely [82Gar].
KWU-Service used ultrasonic decontamination to clean primary recirc pumps at several power plants
(Biblis A-B, Neckarwestheim, Unterweser, Borselle and Atucha). This equipment has also been used in
more recent power plants. At the KWU Centre in Karlstein, Germany, a large amount of scaffolding and
small tools were decontaminated up to the release limit [84KWU].

Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

8 Decontamination

8-21

8.1.3.4.3 Decontamination by mobile ultrasonic tools


A specially designed ultrasonic hand-held wall cleaner and floor cleaner has been designed at the
Argonne National Laboratory in the USA for decontamination of flame-sprayed zinc on hot cell liners
[70Ayr].
8.1.3.5 Decontamination by foams
Foam decontamination uses liquid foam generated from an acid or acid mixture, using air, nitrogen or an
inert gas. The foam also contains various chemical additives such as inhibitors, foam stabilisers and
surfactants [82Com]. Typical acids used are hydrochloric, nitric, hydrofluoric, sulphuric, and phosphoric
and organic acids can also be considered. The foam is produced in a foam generator and the density can
vary considerably typically 1 litre of acid solution is used to produce 20 dm3 of foam. Foams have been
successfully used in decontamination. The use of foams started more than 40 years ago [60Ayr] and at
that time, inhibited hydrochloric acid, together with special surfactants, was used. Decontamination
factors in the range of 5 to 50 are obtained with a single foam application using 7-10 % phosphoric acid
[82Gar].
8.1.3.6 Decontamination by gels
A gel medium is defined as a semisolid system obtained by flocculating and immobilising particles in a
continuous medium. The problem in utilising this method of decontamination is the generation and
maintenance of adequate gel systems in combination with decontaminating chemicals. For example, it has
been found difficult to develop an adequate gel system using alkaline permanganate solutions [82Gar].
Gels can be made from either organic based or inorganic based systems and contain in the gel
formulation decontaminating chemicals which are normally acids such as phosphoric, sulphuric, or nitric
[80Des]. High decontamination factors (in excess of 50) have been obtained in the laboratory for mild
steel, stainless steel, aluminium, copper and Plexiglas. Large decontamination tests by gel spraying
decontamination have been performed at CEA-CEN-Cadarache, France, on 17 Mg of steel from the
German ISAR-BWR (consisting of 11 Mg of frame and 6 Mg of pipes) [89Brun]. The chemicals used
were sulphuric acid (2 mol dm3) and hydrofluoric acid (1.6 mol dm3) and during the decontamination
100 litres of gels were used.
8.1.3.7 Decontamination by pastes
Cleaning pastes are widely used for treating metal surfaces, particularly for stainless steel, and can be
extremely effective for decontamination. They consist of a filler, carrier and use an acid or mixture of
acids as the active agent. The concept of decontamination pastes follows the method used for the older
paste systems, however new acid systems particularly effective in removing radioactive contaminants
have been developed. These pastes, when applied in a thin layer on contaminated surfaces, can provide
effective decontamination, together with generating relatively small quantities of waste.
8.1.3.8 Mechanical decontamination techniques
Mechanical techniques include many decontamination methods based on the use of mechanical tools or
devices to remove the surface contamination [56USS]. Mechanical devices are commonly used for
cleaning industrial tubing and piping and can be adapted for the decontamination of similar items in a
nuclear installation.
Landolt-Brnstein
New Series VIII/4

8-22

8 Decontamination

[Ref. p. 8-34

8.1.3.9 Decontamination by strippable coatings


Decontamination by strippable coatings consists of the application of a coating over the surface to be
decontaminated. This coating is then left on the surface for a set period (from few hours to a few days)
and then removed/stripped resulting in removal of the contamination. Strippable coating formulations
usually consist of high molecular weight, film forming, synthetic polymers such as polyethylene,
polyvinylacetate, polyvinylchloride, acrylics, etc., dispersed as an emulsion in an aqueous base. These
coatings usually contain an active agent e.g., an acid or mixture of acids, which attack the contaminants
on the surface to which the coatings are applied. The coatings may be applied with a brush, spray system,
roller or other similar method. In some cases, it may be necessary to apply two or more coats to ensure
that the coating has sufficient strength to be readily removed from the surface without tearing. The
coatings are applied in varying thicknesses from 0.5 to 2 mm [79TMI]. Usually the coating is then
manually stripped off the surface in sheets, compacted and placed in waste containers.
8.1.3.10 Melting
Melting is considered as a decontamination process since it can be used to reduce the specific
contamination. The method completely destroys components and is effective only for contaminants that
are volatile or more soluble (e.g. plutonium) in the slag than the molten metal. The decontamination
efficiency varies widely depending on the radioisotope present. The radionuclides remaining in the
molten material are distributed homogeneously and effectively immobilised, thus reducing the possibility
of the spread of contamination. The melting should take place in a suitable refinery, which has filters on
the gas exhaust system to protect the environment [85Pfl].
Melting is extensively used in Germany. From 1984 to 1989 more than 2000 Mg of low-level
contaminated scrap (<74 Bqg-1) have been melted and recycled [90Sap].

8.1.4 Decontamination techniques for building surfaces


Decontamination processes to be used for contaminated concrete depend greatly on the characteristics of
the concrete surface to be cleaned. They can vary from very simple hand based processes, to
jackhammers or drilling removal techniques. The former is normally used for cleaning painted or smooth
surfaces covered by loose contamination and the latter for decontaminating concrete in which the
contamination has penetrated deeply. The following techniques a are in use:
Brushing, washing and scrubbing: These are widely and frequently used at nuclear facilities to clean
smooth surfaces, because they are simple and inexpensive. They are generally considered together,
because they are related and in many decontamination works are used jointly or sequentially.
Smearable contamination can be removed by wiping with a dry or damp cloth if the surface is
smooth or impervious. To increase the effectiveness of decontamination detergents and solvents are
added to the solutions especially if the loose contamination is associated with grease or oil. Abrasive
powders or pads can be used if the contamination is associated with rust if it is embedded near the
surface.
Vacuum cleaning: Vacuum cleaning is one of the most widely and frequently used decontamination
processes to clean smooth concrete surfaces. It is also used to collect dust resulting from brushing
decontamination processes such as scarifying, spalling, etc. The process is very Simple and can be
efficiently used for loose particles on both wet and dry surfaces.
Scarifying and grinding techniques: Scarifying and grinding processes [80Bar] have been widely used
for a long time for the decontamination of concrete walls and floors of different nuclear facilities. They
are particularly appropriate for the removal of thin concrete contamination layers (typically less than
10 mm).

Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

8 Decontamination

8-23

Thermal scarifying: Thermal scarifying processes are based on the removal of concrete layer by
thermal energy introduced by flame (or plasma) impingement on the surface to be decontaminated. In
principle two different thermal scarifying phenomena may take part in the process: spattering and melting
[85Ebe].
Both spattering and melting use almost all the thermal energy of the flame, so that no heat penetrates
the material. The energy remains in the spattered and molten material, which becomes separated from the
concrete. After flame scarifying, the loose particles and remnants of combustion must be removed from
the treated surface. Circular brushes or cleaning machines, which can be fitted with steel wire brush
rollers or beater rollers, are recommended for this task. Flame scarifying has long been used to treat
surfaces in order to produce a clean, dry base for paint and other surface coatings.
Spalling: When a floor or wall is deeply contaminated, a thick concrete layer must be removed in order to
decontaminate it completely. Removal of the surface radioactivity in this manner, in comparison with
demolishing the entire structure, eliminates the need to dispose of large quantities of non-radioactive
concrete, which may arise with other volume removal techniques. To remove these thick concrete layers
hard mechanical processes should be used. Surface breakers, pneumatically or hydraulic operated drilled
bits, and water cannons are typically used in spalling processes.

Landolt-Brnstein
New Series VIII/4

8-24

8 Decontamination

[Ref. p. 8-34

8.2 Decontamination of skin


8.2.1 Introduction
Radiation protection rules in national legislation generally include action levels or limits for protection
measures in case of surface contamination at the workplace and of objects (see Section 8.1). Frequently
limits for decontamination measures of skin are not included. However, radiation protection in practice
requires at least reference values to avoid unsuitable or detrimental decontamination measures which may
lead to skin lesions or increased incorporations. Based on new calculations of equivalent dose rates for the
skin [85Hen], recommendations of reference values have been developed, among others, by the German
Commission on Radiological Protection [92SSK] which serve as a basis for the following specifications.
The described measures in case of skin contamination are generally based on the following principle:
In the event of contamination, the resulting radiation dose must be kept as low as reasonably achievable,
economic and social factors being taken into account, and considering individual situations also below the
dose limits for the skin, in accordance with the recommendations of the International Commission on
Radiological Protection ICRP [91ICR] (see also Sect. 4.8).
Adherence to this principle requires in working areas, where contamination cannot be excluded,
suitable measures to keep the skin dose after contamination as low as possible. The following
specifications serve this purpose. They are no recommendations for measures in emergency and disaster
situations and do not include medical treatment of contaminated wounds. They are rather addressed to
technical, medical and scientific installations where sealed and unsealed radioactive sources are handled,
in accordance with the respective radiation protection rules of their national legislation.
The following areas are mainly involved:

Nuclear power plants


Nuclear fuel cycle installations
Scientific and industrial laboratories
Hospitals, medical laboratories and practices of doctors in nuclear medicine.

8.2.2 Transport of radioactive substances via the skin


8.2.2.1 Anatomy of the skin
The skin (cutis) consists of epidermis and subcutis. The epidermis is the avascular external skin layer.
The subcutis consists of a tissue layer with connective tissue septa in which fat cell clusters and nerves
are located.
In case of skin contamination the epidermis is primarily concerned. Due to permeation of radioactive
substances, radioactivity may enter the transfer compartment (see Chapter 7) via the subcutis and thus
lead to internal contamination of the organism.
The epidermis consists of multi-layer corneal squamous-cell epithelium of a thickness between 30 m
and 2 mm, depending on the body region:
The external layer of the body skin, i.e. the upper layer of the epidermis, is the stratum corneum, on
the surface of which flat, denucleated corneal cells peel off in fine scales. It is followed by the stratum
lucidum consisting of denucleated cells which is only produced at thick parts of the epidermis - palm and
sole. It is followed by the stratum granulosum, the so-called granular cell layer. Then follows the stratum
spinosum, the so-called prickle cell layer and the stratum basale, the basal-cell layer. Stratum spinosum
and stratum basale are also called stratum germinativum (germ cell layer), because here the corneal cells
scaled off at the surface of the epidermis are substituted by cell division. Therefore, the cells of the
stratum germinativum are the radiation-sensitive cells of the epidermis. Their radiation dose (equivalent
dose) has to be determined in case of skin contamination, their dose level determines the introduction and
conclusion of decontamination measures.
Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

8 Decontamination

8-25

8.2.2.2 Transport procedure


The detriment to health due to skin contamination from radioactive substances depends mainly on

Type of the radionuclide and its chemical compound


Activity per area and/or specific activity
Solubility of radioactive substance
Situation and size of possibly affected part of skin.

In principle the healthy skin is protected best against percutaneous incorporation of radioactive
substances. Therefore, all persons handling unsealed radioactive sources should give special care to their
skin, particularly at their hands, and keep it in a good and healthy condition, because fissured skin surface
may become openings for entry of radioactive substances and consequently for incorporations of
radionuclides.
The intact skin is an effective but not completely dense barrier against radioactive substances.
Whereas solid particles, preferred by rubbing, may enter the skin mechanically, liquids are subject to
capillary forces and diffusion processes. As long as a liquid wets the skin surface, a transport of material
occurs from the surface into the skin and through the skin. This leads to both a transient deposit of
radioactivity in the corneal layer and transport into deeper skin layers (subcutis) together with uptake into
blood and subsequent internal radiation exposure (see Chapter 7).
The capacity of the corneal skin layer to take up radioactive liquid is exhausted within several
minutes. This layer takes up about one micro litre liquid per square centimeter wet surface [92Pra].
Activity from this procedure taken up into the corneal layer is therefore proportional to the specific
activity of the liquid and to the size of the affected skin surface. However, the corneal layer may have a
special affinity to some substances. In case of a permanent contamination this may lead to radioactive enrichment in the corneal layer.
The transport through the skin (permeation) from a liquid on the surface is proportional to the specific
activity in the liquid and to the size of the affected skin surface and additionally to the time period during
which the contaminated liquid remains on the skin. In case of inorganic substances dissolved in water,
organic acids, salts, or lipophilic compounds, the substance transported through the skin per square
centimeter and hour is equal to the amount contained in 0.001 to 0.1 l [92SSK]. The permeation rate is
significantly higher with gases dissolved in water or easy volatile substances.

8.2.3 Skin dose at contamination


8.2.3.1 Calculation of the equivalent dose to the skin
The equivalent dose of the contaminated skin can be calculated as follows [85Hen]:
ln 2

t
86400
T1/2 &
H S = AF T1/ 2
h =
1 e
s
ln 2

(8.2.1)
ln 2

t
T1/2 &
5

H S = 1.25 10 AF T1/ 2 1 e
h

with HS equivalent dose of contaminated skin in Sv


AF
activity per area at the surface in Bq/cm2
physical half-life in days (d)
T1/2
t
time of contamination in days (d)
h&S
equivalent dose rate conversion coefficient in Sv/s per Bq/cm2 (see Section 8.2.3.2)
Landolt-Brnstein
New Series VIII/4

8-26

8 Decontamination

[Ref. p. 8-34

If a dwell time of one week is assumed for contamination, equation (8.2.1) simplifies for long-lived
radionuclides, i.e. for radionuclides with significantly longer half-life than 7 days, as follows:
HS = 604800 AF h&S

(8.2.2)

8.2.3.2 Equivalent dose rate conversion coefficients

The equivalent dose rate conversion coefficient for skin contamination is defined as follows (see equation
8.2.1):
The numerical value of the equivalent dose rate conversion coefficient corresponds to the equivalent
dose rate in Sv/s in the contaminated skin at an activity per area of 1 Bq/cm2. For determining these
values [85Hen] the mean skin dose was calculated by integration over a skin depth between 50 and 100
m, and the contribution of gamma-, beta-, electron radiation (Auger electrons) and alpha-particles was
considered. Contamination of the whole skin surface was assumed to calculate the contribution of gamma
radiation to skin dose. The assumption that the radiation-sensitive layer (stratum germinativum, Sect.
8.2.2.1) is situated mainly in a skin depth of 50 to 100 m leads to somewhat higher equivalent dose rate
conversion coefficients than those obtained for the reference skin depth of 70 m [91ICR].
Radioactive substances may enter the corneal layer, however, the activity concentration decreases
significantly with depth of the corneal layer (exponentially with a half-value thickness of about 2 m). As
a consequence this permeation has only little influence to dose in the radiation-sensitive skin layer
(stratum germinativum). Therefore the ambient activity distribution in the corneal layer was not
considered in the calculation of the equivalent dose rate conversion coefficents.
The equivalent dose rate conversion coefficient values for 128 radionuclides are summarised in
Table 8.5.
Table 8.5 Equivalent dose rate conversion coefficients (Sv/s/(Bq/cm2)) for contaminated skin (averaged
over a depth of 50-100 m) [85Hen]

Radionuclide

Radiation
Electrons/BetaParticles

Gamma-Radiation

Alpha-Particles

Na-24

4.3E-10

6.6E-11

Cr-51

7.1E-12

1.4E-12

Mn-54
Mn-56

2.1E-12
4.2E-10

1.6E-11
3.0E-11

Fe-55
Fe-59

2.7E-10

1.2E-12
2.2E-11

Co-56
Co-58
Co-60

8.8E-11
7.0E-11
2.4E-10

6.2E-11
1.9E-11
4.5E-11

Ni-59
Ni-65

4.1E-10

1.6E-12
9.7E-12

Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

8 Decontamination

8-27

Radionuclide

Radiation
Gamma-Radiation

Alpha-Particles

Zn-65
Zn-69
Zn-69m

Electrons/BetaParticles
7.5E-12
4.3E-10
2.4E-11

1.3E-11
1.1E-16
7.7E-12

Se-75

3.0E-11

9.3E-12

Rb-86
Rb-88
Rb-89

4.2E-10
1.7E-10
3.9E-10

1.8E-12
1.1E-11
3.6E-11

Sr-89
Sr-90
Sr-91
Sr-92

4.2E-10
3.9E-10
4.2E-10
3.7E-10

1.6E-15
1.3E-11
2.4E-11

Y-90
Y-90m
Y-91
Y-91m
Y-92
Y-93

4.2E-10
5.9E-11
4.2E-10
2.4E-11
3.8E-10
4.1E-10

4.3E-16
1.1E-11
6.6E-14
1.0E-11
4.6E-12
1.5E-12

Zr-93
Zr-95
Zr-97

1.8E-13
2.8E-10
4.2E-10

1.4E-11
3.3E-12

Nb-93m
Nb-95
Nb-95m
Nb-97

4.0E-09
4.5E-10
4.2E-10

4.7E-13
1.4E-11
2.8E-12
1.2E-10

Mo-93
Mo-99
Mo-101

4.1E-10
4.6E-10

2.5E-12
2.9E-11
2.4E-11

Tc-99
Tc-99m
Tc-101

2.6E-10
5.1E-11
4.3E-10

2.2E-12
5.8E-12

Ru-103
Ru-105
Ru-106

1.5E-10
4.2E-10
-

8.8E-12
1.5E-11
-

Rh-103m
Rh-105
Rh-106

3.2E-10
3.9E-10

3.2E-13
1.4E-12
3.8E-12

Landolt-Brnstein
New Series VIII/4

8-28

8 Decontamination

[Ref. p. 8-34

Radionuclide

Radiation
Gamma-Radiation

Alpha-Particles

Ag-110m
Ag-111

Electrons/BetaParticles
1.2E-10
4.2E-10

5.1E-11
4.6E-13

Sb-124
Sb-125
Sb-126
Sb-127
Sb-129
Sb-130
Sb-131

3.7E-10
1.8E-10
3.5E-10
4.1E-10
3.9E-10
4.9E-10
4.1E-10

3.2E-11
8.8E-12
5.3E-11
1.3E-11
2.6E-11
6.0E-11
3.4E-11

Te-125m
Te-127
Te-127m
Te-129
Te-129m
Te-131
Te-131m
Te-132
Te-133
Te-133m
Te-134

2.8E-10
3.9 E-10
1.5E-10
4.4E-10
3.1E-10
4.9E-10
3.9E-10
2.1E-10
4.1E-10
4.4E-10
4.6E-10

3.1E-12
8.9E-14
1.0E-12
1.5E-12
1.3E-12
7.6E-12
2.6E-11
5.0E-12
1.7E-11
4.2E-11
1.7E-11

I-129
I-130
I-131
I-132
I-133
I-134
I-135

9.2E-11
4.1E-10
3.7E-10
4.2E-10
4.2E-10
4.2E-10
4.0E-10

1.7E-12
4.0E-11
6.9E-12
4.2E-11
1.1E-11
4.8E-11
2.8E-11

Cs-134
Cs-134m
Cs-135
Cs-136
Cs-137
Cs-138

2.8E-10
3.3E-10
1.5E-10
3.3E-10
3.7E-10
4.1E-10

2.9E-11
1.4E-12
4.0E-11
4.0E-11

Ba-137m
Ba-139
Ba-140

4.5E-11
4.4E-10
4.1E-10

1.1E-11
7.4E-13
3.9E-12

La-140
La-141
La-142

4.3E-10
4.2E-10
4.1E-10

4.0E-11
7.5E-13
4.4E-11

Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

8 Decontamination

8-29

Radionuclide

Radiation
Gamma-Radiation

Alpha-Particles

Ce-141
Ce-143
Ce-144

Electrons/BetaParticles
4.1E-10
4.3E-10
2.1E-10

1.4E-12
5.7E-11
5.1E-13

Pr-143
Pr-144m
Pr-145

6.6E-13
1.1E-11
4.2E-10

6.7E-12
9.2E-13
2.5E-13

Nd-147

4.0E-10

3.2E-12

Pm-147
Pm-148
Pm-148m
Pm-149
Pm-151

1.3E-10
4.2E-10
3.3E-10
4.2E-10
4.1E-10

1.0E-16
1.0E-11
3.7E-11
1.9E-13
5.7E-12

Eu-152
Eu-152m
Eu-154
Eu-155
Eu-156

2.0E-10
3.3E-10
4.5E-10
8.4E-11
3.7E-10

2.1E-11
5.7E-12
2.3E-11
1.4E-12
2.3E-11

At-211

1.4E-11

1.6E-12

Ra-226

1.1E-11

1.4E-13

U-234
U-235
U-238

8.7E-12
6.1E-11
-

4.6E-13
3.5E-12
3.8E-13

Np-237
Np-238
Np-239

6.2E-11
2.9E-10
6.2E-10

3.1E-12
1.2E-11
4.8E-12

Pu-236
Pu-238
Pu-239
Pu-240
Pu-241
Pu-242

1.0E-11
8.0E-12
1.9E-13
8.4E-12
1.0E-09
7.0E-12

5.3E-13
4.6E-13
2.5E-13
4.4E-13
2.8E-16
3.7E-13

6.9E-20
5.5E-20
1.2E-20
1.3E-20
-

Am-241
Am-242
Am-242m
Am-243

4.7E-11
3.1E-10
2.6E-13
2.1E-11

3.1E-12
1.4E-12
1.0E-12
1.7E-12

5.7E-20
8.1E-20
2.1E-20

Landolt-Brnstein
New Series VIII/4

8-30

8 Decontamination

Radionuclide

Radiation

Cm-242
Cm-243
Cm-244
Cm-245
Cm-246
Cm-247
Cm-248

Electrons/BetaParticles
4.7E-12
3.3E-10
1.2E-10
3.5E-11
-

[Ref. p. 8-34

Gamma-Radiation

Alpha-Particles

4.3E-13
4.2E-12
4.0E-13
3.7E-12
3.5E-13
6.0E-12
2.7E-13

7.7E-15
3.0E-16
8.6E-20
3.0E-20
3.0E-20
4.0E-21
5.4E-21

8.2.4 Decontamination measures


8.2.4.1 Organisational and preliminary measures

When handling radioactive substances contamination should always be anticipated. Therefore,


organisational measures and practical procedures for personal decontamination have to be provided.
There is need for developing special instructions of decontamination for the respective operation and also
for individual workplaces.
These measures include, among others, to take off contaminated clothing before decontamination
measures are started. Care shall be taken that no additional parts of skin will be polluted and no
contaminated dust will be emitted into the air. The emergency staff should wear protective gloves or
protective clothing.
8.2.4.2 First aid measures of skin decontamination

Simple decontamination appliances which can be used immediately after contamination and at any place
should be available for decontamination. If need be, immediate decontamination measures will be given
priority over assessing the value of skin contamination by activity measurement. Based on the experience
that both specific activity concentration of a contaminated liquid and the time of influence or action may
be the decisive parameters for skin permeation, skin decontamination should be started immediately after
contamination, if possible.
However, it should always be considered that incorporation of radionuclides due to permeation might
be effectively reduced by simple and quickly performed washing measures.
In general washing with lukewarm water, special soaps or wash lotions using soft hand-brushes, if
required, are first and rapid decontamination methods. Also secondary contamination of the surrounding
skin from washing procedures with lukewarm water is widely negligible in practice, because if enough
water is immediately used, the radionuclide concentration is significantly decreased and the time of
influence is short.
Minor contaminations can usually be removed already in a first washing course. During this stage
only the contaminated skin parts should possibly be cleaned with lukewarm water, e.g. only the palm of
the hand. Washing should be finished after 2 minutes and the skin should be dried with absorbent
material. In case of remaining contamination the procedure according to Section 8.2.5 should be applied.

Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

8 Decontamination

8-31

8.2.4.3 Specific decontamination procedures


Decontamination by removing contaminated corneal cells

Small-surface contamination can be removed by taking off corneal cells with adhesive film: after 5
strippings 97 % of the radioactive substance was removed from the skin at the forearm [58Bor]. However,
this stripping method is only suitable with dry skin and fails at the palms of the hand.
Cleaning by sorption agents

To remove substances penetrated deeper into the corneal layer, cleaning methods are required where the
procedure of penetration shall be inverse, i.e. from internal to external penetration. In order to avoid
transfer of radioactive substances into the corneal layer from capillary forces, it is suitable to apply for the
decontamination sucking sorption agents, e.g. silicon dioxide, titanium dioxide or silica gel.
Cleaning by detergents

Although cleaning with customary detergents is a natural decontamination method, it should be


considered that without penetration of a decontamination agent into the corneal layer, the substance
deposited there cannot be reached. Therefore when cleaning a substance situated in the corneal layer it
shall be dissolved by the detergent and rinsed to the skin surface. Therefore the corneal layer must not be
decontaminated by alkaline or strong acidic cleaning agents, because the bond capacity of keratin to ions
increases with each deviation from the pH value 4.2, as it has been proved by experimental results with
22
Na and 131I ions in corneocytes [71EI, 84Pra].
Specific decontamination

Specific decontamination methods have been described by Wijker [66Wij]. However, they should only be
performed by specially trained experts. Warning is particularly issued about chemicals for nuclidespecific decontamination, if there is no knowledge about the chemical composition of radionuclides
involved.
Decontamination agents for skin and hair

For the decontamination of skin and hair the following decontamination solutions are normally used:
Decontamination lotions for skin and hair

Titanium oxide paste (general skin decontamination procedure)


Wiping paste (general skin decontamination procedure)
Citric acid 3 % (e.g. decontamination of hair and external auditory canal)
Complexing solutions (e.g. in case of contamination of the eyes, general skin decontamination
procedure) [97Ger]
Potassium permanganate solution (general skin decontamination procedure), removing brown skin
colouring with sodium disulfite solution
Physiological sodium chloride solution (decontamination measure also in case of contamination of the
eyes).

Landolt-Brnstein
New Series VIII/4

8-32

8 Decontamination

[Ref. p. 8-34

8.2.4.4 Decontamination of specific body regions and organs


Hair

Contaminated hair should be washed with a wash lotion (see Section 8.2.4.3), assisted by a helper
wearing protective gloves, in an adequate hair washbasin with the head bent backwards. Then the hair
should be rinsed with plenty of water. Special care must be taken that no contaminated water runs into the
face, eyes or ears. Before hair drying, control measurement by contamination monitor is required.
Eyes

In case of contamination of the eyes, these should be properly rinsed with plenty of water; cleaning
lotions (physiological sodium chloride solutions and integration solutions (see Section 8.2.4.3) should be
used, if necessary. This method of eye decontamination must be performed under medical supervision.
Mouth, nose, ears

If mouth, nasopharynx and auditory canal are contaminated, a physician must always be contacted.
The mouth should be rinsed with plenty of water for decontamination.
Contamination of the nasal cavities can be decreased by blowing the nose.
Rinsing (wash bottle) with physiological sodium chloride solution or citric acid must only be done on
the instructions or assistance of a physician, if possible, because there is hazard of radionuclide
incorporation. In any case secondary contamination has to be avoided.
Skinfolds, groove of the nail bed, and fingernails

If contamination is detected in skinfolds, in the groove of the nail bed or under the fingernails, this should
be specifically removed. Simple instruments can be used such as nail cleaner, soft brush or adhesive
strips.

8.2.5 Procedure at residual contamination and fixing a reference value


8.2.5.1 Frequency of decontamination steps

If the first decontamination procedure (see Sections 8.2.4.2, 8.2.4.3 and 8.2.4.4) is not successful, the
decontamination method can be repeated up to two times while measuring each individual decontamination effect. If the decontamination effect is lower than 10 %, and the surface-related residual
activity is lower than the reference value of 10 Bq/cm2 averaged over 100 cm2 with contamination
predominantly dispersed over the whole surface (see Section 8.2.5.2) the additional decontamination
step can be waived. As far as the decontamination effect is higher than 10 % and the skin condition is
good, further wash procedures may be reasonable.
8.2.5.2 Derivation of the reference value for residual contamination

The reference value of 10 Bq/cm2 of a remaining skin contamination after several decontamination steps
leads to the fact that for more than 90 % of the radionuclides listed in Table 8.5 which are essential for
practical radiological protection purposes the remainder equivalent dose is significantly less than 1 % of
Landolt-Brnstein
New Series VIII/4

Ref. p. 8-34]

8 Decontamination

8-33

the annual dose limit to skin of 500 mSv [91ICR] in a 1-week-dwell time. All dose values lie below 5 %
of this limit, except for 254Cf. This stipulation ensures that even in case of several contamination events in
the calendar year which were not successfully removed, the distance to the limit value is sufficient.
When fixing a reference value to decide on further decontamination measures, it can also be assumed
that activity concentration decreases exponentially with depth in the corneal layer. The complete scaling
off of this layer within two weeks leads to a very quick exponential decrease with time of the residual
activity in the skin. Consequently the 1-week-dwell time taken as a basis for dose calculations (see
equation 8.2.1 in Sect. 8.2.3.1) overestimates significantly the actual equivalent dose.
Table 8.6 gives an example of the activity per area values in some radionuclides relevant for
radiological protection purposes. Considering the physical half-life at 1-week-dwell time they lead to a
dose of about 1 % of the annual dose limit to skin of ICRP of 500 mSv [91ICR].
Table 8.6 Activity per area of some radionuclides leading after 1-week-dwell time to a skin equivalent
dose of about 5 mSv (1 % of the annual dose limit for skin of 500 mSv [91ICR]).
Radionuclide
Activity per area
[Bq/cm2]
14
C
170
60

Co

35

90

Sr

20

90

40

131

35

137

Cs

20

141

Ce

20

The reference value of 10 Bq/cm2 is adequately conservative and can also be used for radionuclides
which, due to the short range of radiation emitted (mainly Auger electrons), provide a main contribution
to dose in the skin layer sensitive to radiation (stratum germinativum). Consequently, precaution against
the hazard of transmitting radioactive substances from the restricted access area is ensured, whereby it
should be taken into account that remaining residual contamination is a very rare event and that remaining
activity clings tightly to the skin.
Apart from exposure to skin, radionuclides situated in the corneal layer may principally lead to
exposure in other body regions:
Radiation mainly gamma radiation may expose other body organs or tissues
Radioactive substances may reach body liquids by permeation and thus disperse in the body with final
irradiation of organs or tissues.
Usually, external exposure to other body regions by gamma radiation resulting from the corneal layer
compared to skin exposure is negligible. In case of very high surface contaminations, however, very large
affected body surfaces and long contamination periods, the permeation of radionuclides through the skin
and hence internal dose to body organs and tissues can play a role that should not be underestimated.

Landolt-Brnstein
New Series VIII/4

8-34

8 Decontamination

8.3 References
56USS
58Bor
60Ayr
64Ame
66Wij
70Ayr
71El
78All
78Rie
79Rem
79TMI
80Bar
80Des

80Man
81McV
81Nel
81Rem
82Com
82Gar
82Gar1
83Eic
83Int
84Ebe
84KWU

Fabrication of USS Stainless Steels, 2nd ed., p 88, Bulletin published by United States Steel
Corporation, Pittsburgh, Pa, 1956.
W. Born: Beseitigung radioaktiver Verunreinigungen von der Haut des Menschen;
Strahlentherapie 106, 435 (1958)
Ayres, J. A., Demmitt, T.F., Larrick, A.P., Neubow, G.E., Richman, R.B., Perrigo, L.D.,
Weed, R.D.: Decontamination studies for HAPO water-cooled reactor systems; USAEC
HW-67937. Dec. 27, 1960.
American Society for Metals: Abrasive blast cleaning. Metals Handbook, 8 ed. Vol. 2, 1964.
Skin Contamination; Euratombericht 41-67 (1966)
Ayres, J.A. (ed.): Decontamination of nuclear reactors and equipmentNew York: . The
Ronald Press Co., 1970.
R. El-Julani: Zur Adsorption von Na+ und I an Keratinzellen der menschlichen Haut; Med.
Dissertation, Ludwig-Maximilians-Universitt Mnchen (1971)
Allen, R.P., Arrowsmith, H.W, Charlot, L.A., Hooper, J.L.: Electropolishing as a
decontamination process: Progress and applications; PNL-SA-6858, April, 1978.
Riess, R., Bertholdt, H.: Chemische Dekontamination von Reaktoranlagen, Reaktortagung
1978, 4.-7. April in Hannover, Tagungsbericht, S. 963-966.
Remark, J.F., Miller, A.D.: Review of plant decontamination methods, Sun Valley (ID),
USA: American Nuclear Society, September 17-19, 1979.
TMI Reports: Evaluation of strippable decon coatings; TMIReports No.'s 1-22 (1979).
Barbier, M.M., Chester, C.V.: Decontamination of large horizontal concrete surfaces
outdoors; CONF-800542-2, ORNL, TN (USA), 1980.
Desryoches, J., Koenig, J., Lebrun, J.C.: La dcontamination en milieu glifie ou colloidal;
Workshop on waste washing; Organised by Radioactive Waste Management Committee and
OCED Nuclear Energy Agency at Centre d'Etudes Nuclaires de Cadarache, France.
November 19-21, 1980.
Manion, W.J., Laguardia, T.S.: Decommissioning Handbook; DOE/EV/10128-1, November
1980.
McVey, J.T., et al.: Tools and equipment: From nuclear waste to reusable items, Nucl. Chem.
Waste Manag. 2-3 (1981).
Nelson, J.L., Divine, J.R.: Decontamination processes for restorative operations and as a
precursor to decommissioning: A literature review. PNL-3706, Battelle-Pacific Northwest
Laboratory, May 1981.
Remark, J.F.: Plant decontamination methods review; EPRI NP-1168; May 1981.
Commissariat lEnergie Atomique, Institut de Protection et de Sret Nuclaire:
Dcontamination radioactive du matriel', Publication PMDS, Mars 1982.
Gardner, H.R., Allen, R.P., Polenz, L.M., Skiens, W.E., Wolf, G. A.: Evaluation of
nonchemical decontamination techniques for use on reactor coolant systems; EPRI NP-2690,
October 1982.
Gardner, H.R., et al.: Comparison of decontamination techniques for reactor coolant system
applications; EPRI NP-2777, December 1982.
Eickelpasch, N., Lasch, M.: Electrochemical decontamination experience at Gundremmingen
power plant; Water chemistry of nuclear reactor, Systems 3, Bournemouth (UK), 17-21
October, 1983.
International Atomic Energy Agency: Decommissioning of nuclear facilities:
Decontamination, disassembly and waste management; Vienna: Technical Report Series No.
230, 1983.
Ebeling, W., Boedeker, B., Rose, K.: Dekontamination von Betonoberflchen durch
Flammstrahlen; EUR 8969, 1984.
KWU Service Report, No. 1, April 1984.
Landolt-Brnstein
New Series VIII/4

8 Decontamination
84Pra
85Duc
85Ebe
85EPR
85EPR1
85EPR2
85EPR3
85Hen
85Lr
85Pas
85Pfl
86EPR
86EPR1
86Gau
86Man
87Pav
88Int
89Brun
89Cos
89Cro
89Sta
89Wil
90Ber
90Sap

8-35

H. G. Pratzel, K. Dirnagel, H. Drexel: Kontamination der menschlichen Haut durch


Radionuklide; Nuklearmedizin 23, 197-200 (1984)
Duce, S.W., Simpson, F.B., Mandler, J.W.: Observations of plant decons; EPRI Seminar
on Chemical Decontamination of BWRs, Charlotte (USA), February 26-28, 1985.
Ebeling, W., Boedeker, B., Rose, K., Schaller, R.H.: Decontamination of concrete, with
particular reference to flame scarifying; Decommissioning of nuclear power plants;
Luxembourg 22-24 May 1984; EUR 9474, 1985.
EPRI: EPRI NP-4222, Vol.2, 1985.
EPRI: EPRI NP-4222, Vol.3, 1985.
EPRI: EPRI NP-4222, Vol.4, 1985.
EPRI: EPRI NP-4222, Vol.5, 1985.
K. Henrichs, C. Eiberweiser, H.G. Paretzke: Dosisfaktoren fr die Kontamination der Haut
und der Kleidung; GSF-Bericht 7/85, 5-1285 (1985)
Lrcher, G., Chapuis, A.M., Essmann, J.: Factors to be considered in deciding whether to
decontaminate for unrestricted release; Decommissioning of nuclear power plants,
Luxembourg. 22-24 May 1984, EUR 9474, 1985.
Pascali, R., Bregani, F., Ahlfnger, W., Lasch, M., Gauchon, J.P.: Chemical and
electrochemical decontamination; Decommissioning of nuclear power plants; Luxembourg,
22-24 May 1984, EUR 9474, 1985.
Pflugrad, K., et al.: Treatment of steel waste arising from decommissioning of nuclear
installations by melting; Bethesda, (MD) USA: Nuclear Reactor Decommissioning Planning,
1985.
EPRI: EPRI NP-4687, 1986.
EPRI: EPRI NP-468, 1986.
Gauchon, J.P., Mordenti, P., Bezia, C., Fuentes, P., Kervegant, Y., Munoz, C., Pierlas, C.:
Decontamination par des methodes chimiques, electrochimiques et au jet d'eau; EUR 10043,
1986.
Mang, M.: Issue of the second seminar on chemical decontamination, BWRs-Section 3,
Corrosion Issues, 1986.
Pavlik, O., Sipos, T., Vicsevne, M., Miko, M.: Decontamination of Nuclear Facilities by
Electrochemical methods; 1987 International Decommissioning Symposium, Pittsburgh,
(PA) USA, October 4-8, 1987.
International Atomic Energy Agency: Decontamination and demolition of concrete in the
decommissioning of nuclear facilities; Vienna: Technical Reports Series No. 286, 1988.
Brunel, G.: Decontamination using chemical gels, electrolytical SWAB, abrasives;
Decommissioning of Nuclear Installations, Bruxelles (B), October 24-27, 1989, EUR 12690,
1990.
Costes, J.R., Antoine, P., Gauchon, J.P.: Decontamination before dismantling a fast breeder
reactor primary cooling system; Decommissioning of Nuclear Installations. Elsevier Science
Publishers Ltd, EUR 12690, 1989, ISBN 1-85166-523-4, p. 554
Crossley, H., Wakefield, J.R.: Development of techniques to decontaminate the WAGR Heat
Exchangers, UK, Windscale, Decommissioning of Nuclear Installations. Elsevier Science
Publishers ltd, EUR 12690, 1989, ISBN 1-85166-523-4, p.567.
Stang, W., Fischer, A., Rubischung, P.: Large-scale application of segmenting and
decontamination techniques, Decommissioning of Nuclear Installations, Bruxelles (B),
October 24-27, 1989, EUR 12690, 1990.
Wille, H., Bertholdt, H.O.: Recent developments in component and system decontamination,
conference on water chemistry of nuclear reactor systems 5. London: BNES, 1989, p. 163.
Bertini, A.: Some remarks about decontamination; Decommissioning of Nuclear
Installations, Bruxelles (B), October 24-27, 1989; EUR 12690, 1990.
Sappok, M., Lukacs, G., Ettemeyer, A., Stang, W.: Melting of radioactive metal scrap from
nuclear installations, Decommissioning of Nuclear Installations, Bruxelles, October 24-27,
1989, EUR 12690, 1990.

Landolt-Brnstein
New Series VIII/4

8-36
91Bru
91ICR
92Com
92Pra
92SSK
97Ger
00Hir
00Hir1
00Kat
00Kat1
00Wil
01Eic

8 Decontamination
Brunel, G., Gauchon, J.-P., Kervegant, Y., Josso, F.: Nouvelles techniques de
dcontamination: Gels chimiques, electrolyse au tampon et abrasivfs; EUR 13497 FR, 1991.
1990 Recommendations of the International Commission on Radiological Protection ICRP
Publ. 60; Annals of the ICRP, 1991, Vol. 21 No. 1-3; Pergamon Press Oxford, New York,
Frankfurt, Seoul, Sydney, Tokyo (1991)
Commission of the European Communities: Ispra, Joint Research Centre, Institute for Safety
Technology, Private Communication, 1992.
H.G. Pratzel: Dekontamination der Haut aus der Sicht experimenteller Ergebnisse; in SSK
Verffentlichungen Band 18; Gustav Fischer Verlag Stuttgart, Jena, New York, 71-95 (1992)
Strahlenschutzkommission SSK: Massnahmen nach Kontamination der Haut mit
radioaktiven Stoffen; Empfehlung der SSK vom 22. September 1989; in SSK
Verffentlichungen Band 18; Gustav Fischer Verlag Stuttgart, Jena, New York, 1-30 (1992)
P. Gerasimo, D. Jourdain, A. Cazoulat, D. Schoulz, P. Laroche, R. Ducousso: Modeling of
cutaneous radio-contamination: effects of washings by soap and by solutions of DTPA (in
French); Ann Pharm. Fr. 55 (3), 116-124 (1997)
Hirabayashi, T., Ishigure, K., et. al.: Handbook of reactor water chemistry, Corona Pub. Co.,
2000, p. 276.
Hirabayashi, T., Ishigure, K., et. al.: Handbook of reactor water chemistry, Corona Pub. Co.,
2000, p. 275.
Kato, S., Ishigure, K., et. al.: Handbook of reactor water chemistry, Corona Pub. Co., 2000,
p. 284.
Kato, S., Ishigure, K., et. al.: Handbook of reactor water chemistry, Corona Pub. Co., 2000,
p. 284.
Wille, H., Bertholdt, H., Lessons, H.O.: Learned in full system decontamination by
application of the CORD family concept, BNES,VIII Int. Conference on Water Chemistry of
Nuclear Reactor Systems, Bournemouth, UK -26.10.2000.
Eickelpasch, N., Steiner, H.: Stilllegung von Kernkraftwerken, VGB Power Tech. 6, 2001, p.
142.

Landolt-Brnstein
New Series VIII/4

Ref. p. 9-31]

9 Decorporation of radionuclides

9-1

9 Decorporation of radionuclides

This Chapter first provides an overview of the factors which influence the treatment of persons internally
contaminated with radionuclides and of the available methods of treatment. However it is devoted mainly
to the decorporation of tritium, strontium and iodine isotopes and the actinides plutonium, americium,
thorium and uranium which continue to be a matter of concern. Important cases published in the scientific
literature are summarised and progress made in research studies designed to optimise treatment for
different chemical forms of the actinides reviewed. The Chapter concludes with priorities for future
research and an extensive bibliography.

List of symbols and abbreviations


a
ALI
Bq
CED
DTPA
EDTA
EHDP
EU
ICRP
ID
ILD
i.p.
i.v.
log
N
ORAU
SD
SE
Sv
TBP

Landolt-Brnstein
New Series VIII/4

Year (annum)
Annual Limit on Intake
Becquerel
Committed Effective Dose
Diethylenetriaminepentaacetic acid
Ethylenediaminetetraacetic acid
Ethane-1-hydroxy-1,1,-biphosphonate
European Union
International Commission on Radiological Protection
Injected Dose
Initial Lung Deposit
Intraperitoneal injection
Intravenous injection
The overall stability constant for a metal-ligand complex or chelate
Number of observations
Oak Ridge Associated Universities
Standard Deviation
Standard Error of the Mean
Sievert
Tri-butylphosphate
Mean (average)

9-2

9 Decorporation of radionuclides

[Ref. p. 9-31

9.1 Introduction
Several useful handbooks and reviews on the decorporation of radionuclides from the body have been
published over the years [78V1, 80N1, 84W1, 92B1, 00H1]. However, in the course of time, views and
opinions change on the need for treatment, the radiation doses at which treatment should be considered or
implemented, the most appropriate substance to be used and the optimum treatment regimen. The purpose
of this Chapter is to review and update these issues with particular emphasis on hydrogen (tritium),
strontium, iodine, caesium, plutonium, americium, thorium and uranium. These elements are amongst
those of most concern as a consequence of accidents and incidents involving radioactive materials.
Priority is given to uptakes resulting from inhalation and wound contamination.
The Chapter commences with an overview of factors that affect the efficacy of treatment, treatment
decisions, decision levels and the perception of risk (Section 9.2). This is followed by summaries of the
various methods of treatment (Section 9.3), the efficacies of chelating agents for different chemical forms
of the actinides (Section 9.4) and recent developments in this field (Section 9.5). Much of the Chapter is
devoted to the most effective treatment regimens for different chemical forms of the elements considered
here, as identified by both human experience and animal studies (Section 9.6). The Chapter concludes
with suggestions for future research (Section 9.7) and a comprehensive bibliography (Section 9.8).

9.2 General considerations


9.2.1 Factors affecting the efficacy of treatment
The efficacy of treatment using chelating agents can be affected by the mode of intake, mass and physicochemical form of the contaminant, the reactions of the radionuclide with biological ligands at the site of
entry in the blood and at the sites of secondary deposition, the absorption kinetics of the radionuclide into
the blood, the method and duration of treatment and the mole ratio of the radionuclide to chelating agent.
In principle, the efficacies of clinically approved chelating agents are best evaluated after accidental
human exposure. In practice this may be difficult for some radionuclides, notably the actinides, owing to
uncertainties in the physico-chemical form, pattern of intake, and assessment of intake. Moreover, the
chelating agent may not have been administered by the most appropriate route or the optimum protocol
adopted. Animal studies, when properly executed, need not suffer these disadvantages and moreover are
likely to be the only effective means for evaluating new substances and protocols.
The method of administration favoured by most physicians is slow intravenous injection or infusion
since it is considered that chelation will be most effective when the radionuclide is present in circulating
blood. In general, this is not true. Many studies with laboratory animals have shown that chelating agents
are most effective for biologically soluble forms of radionuclides when they are present at the site of
deposition, for example in the lungs or at a wound site. In these circumstances, local administration of the
chelate is almost certainly the best option. However, when absorption into the systemic circulation occurs
over an extended period then continual intravenous infusion, either directly or as a consequence of oral
administration may be the most effective regimen. For inhaled biologically insoluble materials,
bronchopulmonary lavage may be the only viable method of treatment (see Sections 2.3.1, 2.4)

Landolt-Brnstein
New Series VIII/4

Ref. p. 9-31]

9 Decorporation of radionuclides

9-3

9.2.2 Factors influencing treatment decisions


The aim of treatment is to reduce the risk of deleterious effects to the patient, usually cancer, by reducing
the radiation dose. However, for some compounds of uranium considerations of chemical toxicity may
over-ride radiotoxicity.
It is most important that the organisation or industry should have a clear policy concerning treatment
and that all personnel are familiar with this policy. This should state that the final responsibility for
treatment must always rest with the physician or appointed doctor. Nevertheless, in formulating policy
several points should be addressed, such as:

Does the maximum credible dose warrant treatment?


What are the uncertainties in the assessment of intake and dose?
Can any intake of sufficient magnitude to require treatment be rapidly confirmed?
Is the material amenable to treatment?
What is the likely reduction in dose?
Has the age and general health of the individual been taken into account?
What is the psychological condition of the individual?
Is the outcome of treatment likely to be beneficial when compared with the potential risks ?

9.2.3 Decision levels


An important aspect of radiological protection for the organisation or industry concerned is that there
should be a clear policy concerning treatment and that all personnel are familiar with this policy. The
International Commission on Radiological Protection (ICRP) in Publication 60 [91I1] advises against the
application of current dose limits for deciding on the need for, or scope of, treatment whilst recognising
that at some level of dose treatment should occur. Hence a clear distinction must be drawn between the
dose limit and decisions concerning treatment.
Nevertheless, in practice, treatment decisions, other than for soluble compounds of uranium, will
usually be related to the effective dose limits recommended by the ICRP, namely 20 mSv a1 averaged
over a defined period of 5 a with a further provision that the dose should not exceed 50 mSv in a single
year [91I1, 96O1]. Since in many countries the annual dose limit is restricted to 20 mSv a1, this value
forms the basis of the decision levels suggested here. It is recognised that there are likely to be differing
views on the magnitude of such decision levels. The recommendations given below are the same as those
given in recently published EU reports on decorporation from the human body [92B1, 00W11].
9.2.3.1 Inhalation
For intakes of biologically soluble material, treatment should not be considered when the assessed dose is
below 20 mSv.
For assessed doses between 20 mSv and 200 mSv, treatment should be considered [92B1, 00W1].
Although clinical effects from the intake are unlikely, psychological factors will probably be important.
Single or short-term administration will usually be sufficient. However, if the assessed dose is greater
than 200 mSv, then extended or protracted treatments should be considered depending on the magnitude
[92B1, 00W1].
For intakes of biologically insoluble material such as 239Pu dioxide, the treatment of choice is bilateral
pulmonary lavage but should only be undertaken if there is a likelihood of deterministic effects [92B1,
00W1]. It has been suggested that lung lavage should be considered only when the estimated lung dose is
Landolt-Brnstein
New Series VIII/4

9-4

9 Decorporation of radionuclides

[Ref. p. 9-31

likely to exceed 5 Sv within a few weeks. It should be noted that whilst this procedure is considered to be
of low risk [95D1], attributable to mortality from general anaesthesia [00W1], the lung content will be
reduced only by about two-fold [89N1].
Biologically soluble compounds of low enriched, depleted or natural uranium are potentially
nephrotoxic. The basis for current limits on intake is a maximum kidney concentration of 3 g g1 [73S2,
96H]. It can be deduced, using the ICRP human respiratory tract model [94ICRP] and the systemic model
for uranium [95ICRP], that this value will be attained after acute inhalation of 30 mg and 230 mg of a
very soluble (Type F) or a moderately soluble (Type M) compound, respectively, of uranium [97S1,
98S2].
9.2.3.2 Wound contamination
After a serious accident involving injury and wound contamination, necessary life-saving procedures
must take precedence over decontamination. For accidents involving biologically soluble forms of
radionuclides, the first approach should be to reduce contamination by copious washing with water.
However, if radioactivity has entered the systemic circulation, similar criteria to those described
previously for inhalation should apply.
For wounds contaminated with insoluble materials, washing with copious amounts of water should
again be considered first. In many cases deposits at wound sites can be removed by surgical excision.
Under these conditions it is considered inappropriate to recommend decision levels since many physicians
would wish, provided there is little risk of functional impairment, to remove the radioactivity until it is
below the limit of detection, perhaps a few tens of Bq or less. When, there is a risk of impairment, a
balanced judgement must be made by the physician, preservation of normal function always being the
primary objective.
9.2.3.3 Ingestion
For radionuclides that are extensively absorbed into the bloodstream such as 3H and 137Cs, the criteria for
treatment should again be the same as for inhaled soluble compounds. For ingested insoluble materials
the dose to the lower intestine may be large, with the possibility of deterministic effects. In these
circumstances the use of cathartic or binding agents to accelerate faecal excretion should be used.

9.2.4 Perception of risk and its implications


Rather than have a series of risk coefficients for different individuals in different circumstances, the most
conservative approach will be considered here, i.e. the risk for members of the public. The probabilities
assumed by ICRP for the risk of radiation induced stochastic effects in members of the public are
illustrated in Table 9.1, which shows that the overall risk of health detriment from stochastic effects will
be 7.3 %/Sv [91I1].
Table 9.1. The nominal probabilities for radiation induced harmful effects (from ICRP Publication 60,
1991); [91I1].
Risk [%/Sievert]
Fatal cancer
Non-fatal cancer
Serious hereditary Total
effects
General public
5.0
1.0
1.3
7.3

Landolt-Brnstein
New Series VIII/4

Ref. p. 9-31]

9 Decorporation of radionuclides

9-5

Since decisions on treatment will be based on the net benefit to the patient, it is more appropriate to
consider the overall risk rather than only the risk from fatal cancers. For the suggested decision levels of
20 mSv and 200 mSv, the overall risks from stochastic effects are 0.15 % (~1 in 700) and 1.5 % (~1 in
70). However, other risks to be addressed include the following.
9.2.4.1 The risk from the administration procedure
The route of administration carrying the most significant risk is intravenous injection, in which an air
embolism, leading to serious cardiovascular or neurological effects, or to death is likely to occur in 1 in
20,000 injections (0.005 %) [00W1]. Minor, and reversible, adverse reactions are known to occur in 1 in
40 injections [00W1]. Clearly the likelihood of adverse effects would be increased by repeated administration.
For treatment of inhaled insoluble materials by whole lung lavage, the risks are considered to be
essentially those of a general anaesthetic- between 1 in 50,000 (0.002 %) and 1 in 200,000 (0.0005 %)
[00W1]. It is considered that this level of risk justifies the use of lavage to reduce potential deterministic
effects, but only for lung doses in excess of 5 Sv [00W1].
9.2.4.2 The risk from adverse effects of the therapeutic agent
This is difficult to quantify, and reference to well defined case histories provides the best information. For
example, other than for uranium, the agent of choice for most actinides is DTPA and the usual human
dosage is 0.5 to 1 g of the calcium or zinc salt. In France, over 500 workers have been given a single dose
by slow intravenous infusion and over 200 workers have received multiple doses of DTPA without
adverse effects [87B1]. The Oak Ridge Associated University (ORAU) Registry reported that between
1958 and 1987, 485 patients received a total of 3,077 dosages of DTPA, about two-thirds of them as the
calcium salt. Minor transient effects were observed in 12 patients, but no serious or long term effects were
reported [87B1]. In the Hanford americium accident, 583 g of DTPA, primarily as the zinc salt were
administered to an individual over a 4-year period without any observed toxic effects [89B1].
9.2.4.3 The reduction in risk from treatment
This is again difficult to quantify since it will depend amongst other things on the biokinetics of different
chemical forms of the radionuclide, the method of administration of the chelating agent, and the
frequency and duration of treatment. The spectrum may range from marginal to almost complete removal
of the radionuclide from the body. In broad terms, the extent of removal will reflect the reduction in risk.
9.2.4.4 The risk to the patient in the absence of treatment
Clearly, the risk coefficients for health detriment and the risks associated with the various treatment
procedures referred to above will affect the decision making process. It should be noted that when the
estimated doses are less than about 20 mSv, the risk of treatment may surpass the anticipated benefit.

9.2.5 Approaches to treatment


In broad terms, there are two alternative approaches on which treatment decisions are based. For
convenience these are referred to here as urgent and precautionary.
Landolt-Brnstein
New Series VIII/4

9-6

9 Decorporation of radionuclides

[Ref. p. 9-31

The urgent approach is advisable when a potentially serious intake is suspected but which would take
time to confirm. In this approach the chelating or complexing agent should be administered as a single
dose as soon as possible. The desirability of further administrations would be decided when additional
information on the physico-chemical form of the contaminant, individual monitoring data (whole-body
monitoring or bioassay data as appropriate) or the psychological reaction of the patient becomes
available. The advantage of this approach is that if a high uptake is confirmed, therapy, at least in most
cases, will have commenced at the optimum time. The disadvantage is that if the uptake was not
confirmed, or was trivial, or the material was not amenable to effective chelation therapy, then the patient
might have been subject to an unnecessary, albeit small, risk. It should also be remembered that even the
single administration of a chelating agent may substantially delay the accurate assessment of uptake,
particularly if this is from excretion monitoring, and may increase stress to the patient. On the other hand,
treatment often reduces stress to the patient.
In the precautionary approach, treatment is withheld until uptake is confirmed. The decision to treat
can then be based on the likely magnitude of the uptake and probable reduction in risks of late effects.
Although confirmation of small uptakes can take some time, the advantage of the precautionary approach
is that should the intake be unconfirmed, or assessed as low, then any risks associated with treatment will
have been avoided. The disadvantage of this approach is that should the estimated uptake be above the
decision level, then the efficacy of treatment is likely to be reduced appreciably.
Other than for lavage, which can be delayed for a few weeks without reduced effectiveness, the
authors do not favour one approach over another. They leave any decisions to the professional judgement
of the physician and radiological health-team who will have considered all the options based on local
knowledge.

9.3 Methods of treatment


This Section considers in some detail the different treatment regimens suitable for removing radionuclides
from the body, but with emphasis on tritium, iodine, strontium, caesium, and the actinides plutonium,
americium, thorium and uranium.

9.3.1 Non-specific procedures


These procedures can be applied to any radionuclide and any radioactive compound. They include gastric
lavage to remove material from the stomach; copious washing of a wound; the administration of laxatives
for cleansing the gastrointestinal tract; surgical incision for removing material from a wound; and
pulmonary lavage for removing insoluble material from the lungs.
9.3.1.1 Removal from the gastrointestinal tract
Non-specific procedures can be effective when used immediately after ingestion of radioactive material
which is rapidly absorbed from the gastrointestinal tract or which may result in a high dose to the
intestine.
Orally administered antacids or adsorbents are useful for reducing the uptake of soluble forms of
radionuclides from the gastrointestinal tract. The substances recommended have been used frequently in
clinical practice and they represent virtually no risk to the patient after short- term administration.
Suitable laxatives, such as sodium or magnesium sulphate, will be desirable for reducing irradiation of the
lower large intestine irrespective of the chemical form. Enemas or colonic irrigation may also be used for
the same purpose.
Landolt-Brnstein
New Series VIII/4

Ref. p. 9-31]

9 Decorporation of radionuclides

9-7

Specifically, aluminium phosphate or aluminium hydroxide is suitable for strontium (and barium and
radium) [69S2, 92B1], whilst Prussian Blue (ferric ferrocyanide) [Fe4(Fe(CN6)3] will bind caesium (and
rubidium and thallium) in the gut by ion exchange. Prussian Blue thus renders caesium insoluble in the
intestinal lumen and prevents initial absorption from the gut. By breaking the secretionreabsorption
cycle, its continuous administration will reduce appreciably the systemic content of the element. At the
recommended human dosage, usually 3 g d1, Prussian Blue has no known toxicity [92B1]. The oral
administration of alginate has been investigated for strontium [64W1, 67H1 68C1], barium [72H2] and
radium [72V1].
9.3.1.2 Lung lavage
Lung lavage is used to remove alveolar macrophages from the lungs in which particulate material is
entrained. During a lavage procedure, both lungs will be treated alternately under a single general
anaesthesia with multiple washes of warm isotonic saline whilst oxygen is administered to the other lung.
The procedure can be repeated if necessary after 3-4 days. The technique should not be performed before
the particles in the upper airways have been cleared naturally, but it remains a viable option up to several
weeks after exposure. However the total amount of material which can be removed from the lungs does
not generally exceed 50 % [89N1]. The risk associated with lavage is mainly that of general anaesthesia.
However, it has been suggested that it should only be used on healthy people and where the radiation dose
over a period of a few weeks is likely to exceed 5 Sv [00W1].

9.3.2 Procedures to enhance systemic radionuclide excretion


As indicated previously the procedure of choice will be determined by the biokinetic behaviour of the
contaminant and the different mechanisms by which excretion can be enhanced e.g. by the use of diluting,
immobilising or chelating agents.
9.3.2.1 Diluting and immobilising agents
An important example of a diluting agent is the enhancement of tritiated water excretion by means of
forced fluid intake, often in combination with a diuretic, under medical supervision. In these
circumstances, the biological half-time of 3H in the body, usually about 10 d, can be reduced by about
two-fold during the period of treatment; the reduction in the committed effective dose is somewhat less
due to the short period of treatment [71L1, 72H1, 86L1].
Another important example is the reduced deposition of iodine in the thyroid by the administration of
stable iodide or iodate immediately after intake of the radio-isotope.
The most effective treatment regimen for systemic radio-strontium appears to be the prompt
intravenous or oral administration of stable alkaline earth metal salts, usually as their gluconates [80N1,
84W1, 92B1]. Similarly, attempts can be made to dilute, and hence reduce the systemic deposition of
cobalt and radium isotopes by the administration of stable isotopes, or analogous elements.
At present the best available treatment for systemic caesium is the oral administration of Prussian
Blue, which immobilises the element in the gastrointestinal tract (see 9.3.1.1). However, even with
extended administration the reduction in dose is likely to be only about 2 to 3 fold [94M1, 98I1].

Landolt-Brnstein
New Series VIII/4

9-8

9 Decorporation of radionuclides

[Ref. p. 9-31

9.3.2.2 Chelating agents


The formation of radionuclide complexes in the body that lead to their excretion via the kidneys and
urine, and/or liver and faeces is the most appropriate procedure for many radioactive heavy metals, in
particular the lanthanides and actinides. The chelates most widely used for enhancing the excretion of
plutonium, americium and thorium isotopes are the trisodium calcium or zinc salts of
diethylaminetriaminepenta-acetic acid, referred to hereafter as CaDTPA and ZnDTPA. The former is
normally used for initial and single administration, but since it can remove the essential biometals iron,
manganese and zinc from the body, the zinc salt is preferred for extended or protracted administration.
The mode of action of DTPA is the formation of chemically stable complexes of radionuclides in the
extracellular fluids, most potently lung fluid and blood, that are rapidly excreted in the urine and, to a
lesser extent, the faeces without being reabsorbed. DTPA is normally administered by slow intravenous
infusion or injection at dosages of 15-30 mol kg1 body mass (0.5-1 g for a body mass of 70 kg).
Alternatively, it can be administered as an aerosol or orally, usually at a similar dosage. For wound
contamination, local infiltration of the substance is likely to be most effective, but because severe pain is
likely to be associated with the intramuscular injection of the DTPA, a local anaesthetic, e.g procaine
should be added to the solution. No serious side effects have been observed in humans treated with DTPA
[87B1, 89B1, 98 G1] (see Section 9.2.4).
There is no evidence that DTPA is effective for significantly enhancing the removal of uranium from
the body. In some guidebooks, the recommended agent is sodium bicarbonate. However, this is not
supported by controlled studies with laboratory animals under realistic conditions e.g. with delays
between exposure and treatment of 30 min or more.
Chelation therapy is not an option for the alkaline earth elements strontium, barium or radium since
EDTA (ethylenediaminetetraacetic acid), DTPA, and most other chelators, form stronger complexes with
calcium, than with strontium, barium or radium. Thus calcium will be complexed preferentially, and no
useful enhancement of the excretion of the other alkaline earth metals will be achieved. This point is
illustrated by the stability constants given in Table 9.2. The stabilities are expressed as the overall
constant which is the product of the formation constants for each of the individual metal-ligand
reactions involved in the formation of the chelate of interest. For convenience the values are given as log
, the negative logarithm of the constant.
Table 9.2. Stability constants log (see text) for the complexes between the alkaline earth metals and
EDTA and DTPA [74M1]
Element
log
EDTA
DTPA
Ca
10.69
10.83
Sr
8.73
9.77
Ba
7.86
8.78
Ra
7.1
[7.9]*
* Estimated value.

9.4 General comments on the efficiacy of chelating agents for the


actinides
In general, the greatest problems posed in decorporation of radionuclides from the human body involve
the actinide elements. Most of the research conducted in the last decade or so has also concentrated on
these elements. This Section concerns the authors responses to some of the most frequently asked
questions on the efficacy of chelating agents for the actinides.
Landolt-Brnstein
New Series VIII/4

Ref. p. 9-31]

9 Decorporation of radionuclides

9-9

9.4.1 What are the factors that govern the efficacy of chelating agents ?
The efficacy of treatment can be influenced by the mode of intake, mass and physico-chemical form of
the contaminant, the reactions of the radionuclide with biological ligands at the site of entry, the
absorption kinetics of the radionuclide into the blood, the method and duration of treatment, the formation
constant of the metal-ligand complex and the ligand-metal mole ratio.
The mass of the material deposited in the respiratory tract or at a wound site is an important
consideration for predicting the likely efficacy of treatment in human beings and for designing animal
studies. This is particularly relevant for plutonium, americium and thorium which hydrolyse readily at
physiological pH, but will also be important for uranium which can precipitate as phosphate in the lungs.
In animal studies, 238Pu is used frequently for providing mass concentrations of Pu in the respiratory tract
which simulate human exposures to 239Pu (see Section 9.6) more realistically.
Clearly the physico-chemical form will also dictate the availability of the radionuclide to react with
the chelator and thus to enhance excretion. For different materials, the efficacy may be influenced by the
ultrafine component, the rate of dissolution of the particles in-vivo, the reaction of the radionuclide with
biological ligands at the site of deposition and in systemic tissues, its rate of absorption into the blood and
the tissue distribution. The influence of some of these factors are described in more detail in Section 9.6.
The overall efficacy of treatment will also be influenced by the mode of intake of the radionuclide and
the chelating or complexing agent. Invariably, the most likely routes of internal contamination result from
inhalation and wound contamination. After inhalation, chelating or complexing agents could in principle
be administered as an aerosol, by intravenous injection or infusion, or orally. The choice between the
methods will depend on the biokinetics of the contaminant, and whether the substance will cross the airblood barrier or gut wall in sufficient amounts. After wound contamination, chelating agents could be
administered by intravenous injection or infusion, or by local injection. The data obtained from animal
studies suggests that local injection is the preferred method (see Section 9.6)

9.4.2 Can the efficacy of treatment be predicted from animal studies ?


Yes, provided the aerosol characteristics, the mass concentrations of the appropriate chemical forms at the
site of deposition and the mode of uptake represent a realistic accident scenario. In many cases this may
involve the use in animals of a higher specific activity isotope e.g. 238Pu rather than 239Pu. Any
conclusions reached purely on the basis of intravenous injection experiments should be treated with
caution. The differences in the distribution pattern of the radionuclide between species and in their
absorption rates to blood should also be recognised. For example, after the inhalation of a moderately
absorbed compound such as plutonium nitrate, the retention half-time of plutonium would be greater in
the human lungs than in the rat lungs and the fraction of absorbed plutonium deposited in the human
skeleton would be less than in the rat skeleton. On both counts, the overall efficacy of say DTPA would
be expected to be higher in the human than in the rat (see Section 9.6).

9.4.3 Are chelating agents always most effective when the radionuclides are
present in circulating blood ?
No! Ultimately the efficacy of the chelate will be influenced by the biokinetics of the contaminant, and
this should be taken account of in designing the treatment regimen. Often, for radionuclides that are
biologically soluble such as plutonium and americium nitrate, chelating agents will be most effective
when they are deposited at the same site as the contaminant, e.g. in the lungs or at a wound site. However,
for radionuclides which are absorbed into the blood at a moderate rate over a period of time, such as with
238
PuO2 and 241AmO2 after inhalation, then it may be more productive to complex the radionuclide in the
blood so as to prevent its deposition in systemic tissues such as liver and bone. This may require the
continual infusion of the chelate, or its oral administration in drinking water over weeks or months.
Landolt-Brnstein
New Series VIII/4

9-10

9 Decorporation of radionuclides

[Ref. p. 9-31

9.4.4 Is DTPA effective for all actinides ?


No! The major successes in animal studies have been with biologically soluble or moderately soluble
forms of plutonium and americium referred to above. However at present the administration of DTPA
cannot be considered an effective method of treatment for soluble thorium, uranium and neptunium
compounds after inhalation or wound contamination under realistic conditions [00S1, 00S2].

9.4.5 Will the administration of chelating agents result in enhanced tissue


deposition ?
There appears to be no evidence from either human or animal studies that this is an important
consideration when DTPA is used for the decorporation of plutonium and other actinides. However this
may not be true for other chelators, particularly when they are unstable at physiological pH. For example,
research studies with the siderophore analogue 3,4,3-LICAM(C) indicated enhancement of plutonium
deposition in the kidneys [89S2, 89D2], whilst some phosphonates increase substantially the deposition of
uranium in the liver [98H1].

9.4.6 Is the administration of sodium carbonate effective for uranium ?


The administration of sodium bicarbonate has been recommended in various guidebooks and handbooks
for the decorporation of uranium [80N1, 84W1, 92B1]. The evidence available suggests it is not effective,
and in view of the possible side effects such as hypokalaemia and respiratory acidosis its use should be
re-considered for human treatment. Whilst alternative substances such as tiron and some polyphosphonic
acids and the siderophore analogue 3,4,3-LI (1,2-HOPO) have been suggested, the experimental data
show that apart from instantaneous administration they are only partially effective, and usually high
dosages are required [98H1]. The effective decorporation of uranium remains an important problem in
radiological protection.

9.4.7 Must chelating agents be administered promptly to be effective ?


In most cases, yes! It is particularly effective for soluble compounds of plutonium and americium
deposited in the lungs or at wound sites [00S2, Section 9.6]. Prompt administration will minimise
deposition in systemic tissues such as liver and bone from where appreciable removal is exceedingly
difficult. However, the lung content can still be reduced appreciably should treatment be delayed for
several days, although the evidence available suggests that this is not true for wounds [00S2, Section 9.6].
For other chemical forms of plutonium and americium such as 238PuO2 and 241AmO2, prompt
administration will have little effect, and extended treatment as described above, and in Section 9.6 will
be more appropriate.
If effective chelators were available then prompt treatment is essential for soluble uranium compounds
after inhalation and wound contamination in order to mimimise the nephrotoxic effect.

Landolt-Brnstein
New Series VIII/4

Ref. p. 9-31]

9 Decorporation of radionuclides

9-11

9.4.8 Is intravenous injection the best mode of administration ?


This method, favoured by many clinicians, is the means by which most chelating agents can be
administered rapidly. Whilst, in principle, an even more rapid response and greater efficacy (Section 9.6),
could probably be achieved with an aerosol form self-administered with a spinhaler, there is some doubt
about the extent of aerosol deposition in the lungs, and the procedure may be in contravention of the
standing instructions of the employing organisation which would require that a medical officer
administers the substance. However in accidents involving wound contamination, or after intakes
requiring the extended administration of chelates, then intravenous injection would not be the most
appropriate method, and in such cases local and oral administration respectively would need to be
considered.

9.4.9 How can judgements on efficacy be made ?


In the emergency planning stage by consulting the scientific literature on the biokinetics of the same or
similar material to ascertain the absorption kinetics, and whether human or animal data are available to
indicate the likely efficacy of treatment and the optimum treatment regimen. After the accident from
assessments of intake, and retention and excretion data using the most appropriate methods e.g. chest
monitoring, wound monitoring, bioassay.

9.4.10 When should treatment start ?


This will depend on the biokinetics of the contaminant. In most cases, whatever the radionuclide and the
route of contamination, treatment should begin as soon as possible. Delays of a few days would be
appropriate if lung lavage is considered an option (Section 9.2), and might not be critical for compounds
such as 241AmO2 which dissolve fairly slowly in the lungs (see Section 9.6).

9.4.11 When should treatment stop ?


Several criteria are possible. One, when it is evident that the excretion of the contaminant is low
compared with that expected from the estimated internal deposit. This judgement may not be
straightforward. For example, a substantial increase in the urinary excretion rate above background may
still represent a very small fraction of the uptake. On the other hand, an apparent lack of early success
does not preclude the effectiveness of extended therapy, as described for 241AmO2 (Section 9.6). Two,
when the dose or risk has been reduced to an acceptable level (Section 9.2), taking account of the
psychological needs of the patient.

9.4.12 For which materials are chelating agents likely to be effective ?


In the context of this Section, effective treatment implies that the reduction in the committed effective
dose is likely to be at least two-fold, and hopefully much greater. Judgements on the efficacy of treatment
can be based on the published biokinetic behaviour of known chemical forms in animals, and knowledge
of the treatment regimens which can be effective for known soluble or moderately soluble forms; these
Landolt-Brnstein
New Series VIII/4

9-12

9 Decorporation of radionuclides

[Ref. p. 9-31

are described in more detail in Section 9.6. Whilst ligands currently approved for human use must be of
prime consideration, account should be taken of experimental studies with new substances which may
appear to be appreciably superior (Section 9.6)
Provided the criteria affecting decision levels are met (Section 9.2.3), treatment is expected to be
effective for 238Pu and 239Pu inhaled or deposited in wounds as a pure chemical form, e.g. nitrate or
tributylphosphate, provided that treatment commences early and continues for a few weeks (see
Section 9.6)
239Pu inhaled as an oxide with a large ultrafine component (ca 50 % by activity), such as Pu-Na mixed
oxide aerosols [78S1, 79S1 80M1]
238PuO2, provided treatment is extended over many months in order to chelate the Pu arising from the
dissolution of fragmented particles [80 S1, 83M1]
241Am inhaled or deposited in wounds as a pure chemical form of the nitrate, provided treatment
commences early and continues for a few weeks ( see Section 9.6)
241Am present in residues resulting from the refining of Pu metal, provided treatment continues for a
few weeks [87S1]
228Th inhaled or deposited in wound sites as the nitrate, provided treatment commences promptly and
continues for a few weeks (see Section 9.6)
In principle inhalation and wound contamination of 237Np and 239Np. However no effective clinically
approved substance is currently available [00S1]
In principle, uranium inhaled or deposited in wounds as ammonium diuranate, trioxide, nitrate,
tributylphosphate, hexafluoride and tetrafluoride, octoxide and dioxide inhaled as ultrafine particles.
However no effective clinically approved substance is currently available [00S1]

9.4.13 For which materials are chelating agents unlikely to be effective ?


Based on extensive biokinetic studies in laboratory animals to which the appropriate literature references
are given, it is considered that treatment is unlikely to be effective for

238

Pu and 239Pu nitrate intermixed with corrosion products or building dust [87S1, 94M2]
PuO2+ 241AmO2 formed by calcination at high temperatures where 241Am is present as a decay
product of 241Pu [87S1, 95S1]
239
Pu present in residues resulting from the refining of plutonium metal [87S1]
239
Pu present in residues arising from the corrosion of magnox fuel [89S1]
232
Th nitrate , fluoride, hydroxide or dioxide [00S2, 93M1]
237
Np or 239Np dioxide [96I1]
uranium octoxide or dioxide, unless there is a substantial ultrafine component [94A1, 95S1, 96I1,
98A1]
For other materials of potential concern it is recommended that further research on the absorption
kinetics in laboratory animals is undertaken in order to make judgements of the likely efficacy of
treatment.
239

9.4.14 Is lung lavage more effective than chelation treatment for inhaled materials ?
Only if they are essentially insoluble, or dissolve slowly in the lungs over a long period of time. However,
it should be remembered that at best, lung lavage will remove about one half of the radioactivity. The
method would be inappropriate for biologically soluble forms of plutonium and americium. However for
high uptakes of thorium nitrate and 241AmO2, the choice between chelation treatment and lavage is more
difficult to determine.
Landolt-Brnstein
New Series VIII/4

Ref. p. 9-31]

9 Decorporation of radionuclides

9-13

9.5 Recent developments


No significant developments in enhancing the excretion of tritium, radiostrontium, radioiodine or caesium
from the body have taken place for many years. For 3H and radioiodine, it is difficult to envisage how
those procedures in current use can be improved.
Recent developments in decorporation therapy have focussed primarily on the actinides. This is
because the efficacy of DTPA for a variety of chemical forms of plutonium, americium and thorium taken
into the body by various routes has not been fully examined, or treatment using the usual route of intake,
intravenous infusion, has not been completely effective. In addition, no effective agent for uranium
appeared to be available. All this work has been reviewed in more detail elsewhere [ 94S1, 98S1, 98 S2,
00S1,00S2]

9.5.1 Plutonium and americium


It has been recognised for many years that anologues of siderophores were likely to be more effective
than DTPA. Siderophores are sequestering agents produced by microorganisms in order to obtain Fe(III)
from their environment. The basis for this approach was that since the biokinetics of the actinides in
mammals are associated with the Fe(III) transport and storage systems, then the formation constants of
the actinide complexes with siderophore derivatives would be much higher than with DTPA. This was
subsequently found to be the case. Many siderophore analogues such as a linear catechoyl amide codenamed 3,4,3-LICAM(C), a dihydroxamic derivative of DTPA, DTPA-DX, a hydroxypyridinone
derivative of desferrioxamine, DFO-HOPO, a hydroxypyridonate code-named 3,4,3-LI(1,2-HOPO) and
ligands containing the isomer 3,2-HOPO have been synthesised and tested for the decorporation of
plutonium and americium, usually after their intravenous injection as citrates [98D1]. However, animal
experiments involving inhalation and simulated wound contamination using different chemical forms of
these elements, and administration of the ligand by different routes, showed repeatedly that 3,4,3-LI(1,2HOPO) was substantially superior to DTPA (see Section 9.6). Whilst in the early stages the synthesis of
3,4,3-LI(1,2-HOPO) was difficult and expensive, this difficulty has now been largely overcome [98B1].
However it has not yet been approved for human use and studies on optimising treatment for different
chemical forms of plutonium and americium with DTPA remain an important aspect of decorporation
therapy.

9.5.2 Thorium
Studies with rats have shown that DTPA is ineffective for 232Th when deposited as nitrate in the lungs in
amounts that correspond to the annual dose limit for workers. At low masses of thorium, as 234Th, DTPA
is moderately effective after simulating wound contamination by subcutaneous or intramuscular injection,
provided it is administered within minutes. However the most effective ligand developed so far for the
decorporation of thorium after inhalation and wound contamination is 3,4,3-LI(1,2-HOPO). More
information on the most effective treatment protocols is given in Section 9.6.

9.5.3 Uranium
Soluble compounds of uranium are nephrotoxic [89D1, 89L1]. Hence it is important that treatment should
be prompt and effective. Several substances have been investigated in animals. These have included
phenolic compounds such as Tiron, polyaminophosphonic acids, bisphosphonates, and
phosphoalkylphosphinates, and more recently the siderophore analogues 3,4,3-LI(1,2-HOPO) and
Landolt-Brnstein
New Series VIII/4

9-14

9 Decorporation of radionuclides

[Ref. p. 9-31

4-LI(Me-3,2-HOPO). Some of these compounds can reduce the kidney content by about an order of
magnitude compared with untreated animals when the uranium and ligand are administered almost
simultaneously, or within a few minutes. However the efficacy falls sharply with any delay in
administration and they are ineffective beyond about 30 minutes post exposure.

9.6 Optimum treatment protocols


This Section reviews human data where sufficient good quality information is available. However
important data are often obtained from controlled studies with laboratory animals. This procedure is
particularly useful since more than one regimen can be compared for the same exposure scenario, and
new agents can be compared directly with the current clinically approved substance.
The emphasis is placed on inhalation, wound contamination and ingestion. Whilst intravenous
injection of both radionuclides and ligands are used widely in the testing of new substances in animals,
this route of contamination is not important from the standpoint of accidental exposure. However, it is
considered here when data on the other modes of intake are unavailable. In addition it should be borne in
mind that the high efficacy of a ligand observed after intravenous injection of a radionuclide does not
necessarily mean that this will be the case after inhalation or wound contamination. Conversely, the
efficacy may be higher after these routes of intake than after intravenous injection of the radionuclide.

9.6.1 Tritium
9.6.1.1 Human data
Incidents involving the uptake of substantial amounts of HTO are rare. The following case is included
because it resulted in a substantial intake, about 35 GBq, and the treatment regimen used represents the
optimum that can be achieved in practice [86L1]. The individual was encouraged to increase fluid intake
soon after the accident and under medical supervision in hospital, forced diuresis was commenced 100 h
after the accident and continued for 4 d. Diuresis was induced by an intravenous infusion of 7 litres per
day, alternating 1 litre of isotonic saline with 1 litre of 5 % glucose, both being supplemented by 20 mM
of potassium chloride, and further enhanced by giving 40 mg furosimide intravenously each day. In the
4 d of diuresis, which was considered the maximum medically justifiable, 15 GBq of 3H was excreted in
the urine. It was calculated that the above treatment regimen reduced the radiation dose from 800 mSv to
470 mSv. Had forced diuresis commenced immediately after the accident, the dose would be about 410
mSv. Thus for treatment of incorporated 3H, the maximum reduction in dose is unlikely to be more than
two-fold [86L1].
The authors are unaware of any definitive human or animal data on other chemical forms of tritium.

9.6.2 The alkaline earth elements, strontium, barium and radium


The substances recommended for minimising uptake from the human gastrointestinal tract, and hence
systemic deposition, are strontium gluconate (isotope dilution), barium sulphate (insoluble sulphate by
ion exchange), magnesium sulphate (laxative), colloidal aluminium phosphate (antacid) [80N1, 84W1,
92B1].

Landolt-Brnstein
New Series VIII/4

Ref. p. 9-31]

9 Decorporation of radionuclides

9-15

A number of animal studies showed that sodium alginate when given simultaneously or immediately
after oral intake of strontium (or baium or radium) was able to reduce intestinal absorption and thus
retention of the element [72H2, 72H3, 71V1, 77V1, 78V1, 80K1] with little effect on calcium absorption.
A similar effect in humans was reported by Hesp and Ramsbottom [65H1].
Much attention has been paid to methods for enhancing the natural excretion of 90Sr from the body,
these include the administration of diuretics, hormones and the administration of a variety of complexing
agents [68V1, 68W1, 68S1]. However, none of these have suggested a clinically useful procedure and it
is difficult to conceive how the efficacy of removing radiostrontium from the body can be improved with
currently available agents and approaches. Some of the substances tested are given in Table 9.3.
Table 9.3. Compounds which have been
animals or humans
Substance
Reference
Calcium gluconate
68S2, 68V1
Strontium gluconate
68S2, 68V1
Ammonium chloride
68S1, 68S2
Citrate
68S1
Polyphosphates
68S1
Fluoride
68S1
Salicylate
68S1
Phytate
68S1
Alginate
68S1

investigated for the ability to mobilize radiostrontium in


Substance
Pilocarpine
Parathyroid hormone
L-Triiodothyronine
Oestradiol
Hydrocortisone
Alginate
Chlorothiazid (Saluric)
Mercurihydrin

Reference
68S1, 68S2
68S1
68W1
68W1
68W1
65H1, 68H1, 71V1
68S2
68S2

9.6.2.1 Human data


In human volunteer studies Spencer et al [67S1, 67S2, 69S2] found that aluminium phosphate effectively
inhibited the absorption of radiostrontium from the human gastrointestinal tract.
Aluminium phosphate is used clinically for the treatment of colitis, however, the recommended
method of administration is by enema and the only listed pharmaceutical preparation is an aqueous
solution containing 6.5 % AlPO4 (Phosphalugel-Klys 00R1). For the immediate treatment of an
accidental oral intake of radiostrontium or radium an appropriate volume of this solution could, with
caution, be administered orally.
In one study [69S2], a single oral dose of aluminium phosphate gel ranging from 300 ml to 100 ml
(100 ml contained 886 mg aluminium and 1016 mg phosphate) was administered to 12 healthy adults
immediately before an oral administration of 85Sr half-way through breakfast. The amount absorbed,
3.60.5 % (mean se) was substantially less than in untreated controls, 28.81.9 %. In the same study
using 9 volunteers, the amount of 45Ca absorbed in treated and control volunteers was much higher and
more variable, 27.03.0 % and 45.04.2 % respectively. Increasing the dosage of the gel from 100 ml to
300 ml had little effect on absorption in either case.
Vanderborght et al [72V1] administered 15 g sodium alginate per day in bread to a woman who had
been contaminated accidentally with 226RaSO4. The faecal excretion appeared to be enhanced for several
days, suggesting that absorption of the isotope from the intestine would have been reduced. However, this
was a single case and any alginate-induced increase in faecal excretion could only be deduced by
comparison with published data from persons contaminated under different circumstances.
Sodium alginate is a polysaccharide isolated from seaweed and containing guluronic and mannuronic
acid residues; it forms very viscous solutions and is not easy to administer in the required amounts. For
many of the studies reported the alginate was incorporated into bread, [71V1] at a level of 5 % alginate.
However, today a number of alginate-containing pharmaceutical preparations are licensed for human use;
for example Gaviscon, is available as tablets containing 500 mg alginate, 100 mg aluminium hydroxide,
25 mg magnesium trisilicate and 170 mg sodium bicarbonate. Such preparations could be used safely for
the immediate treatment of an oral intake of a strontium, barium or radium.
Landolt-Brnstein
New Series VIII/4

9-16

9 Decorporation of radionuclides

[Ref. p. 9-31

9.6.2.2 Animal data


Humphreys et al [72H1] studied the effect of a single feed of bread containing 5 % of different alginates
on the whole-body retention of orally administered 47Ca, 85Sr, 133Ba and 226Ra in mice. Sodium alginate
had no significant effect on the retention of 47Ca, but that of 85Sr, 133Ba and 226Ra were reduced by factors
of 5, 9 and 9, respectively. Harrison [68H1] also reported that the addition of 10 % of sodium alginate to
the diet decreased the absorption of orally administered 85Sr in rats by factors of 4-5 without influencing
the absorption of 45Ca. Kestens et al [80K1] fed sodium alginate containing bread daily to mice for 3
months following intraperitoneal injection of 226Ra and studied the retention of the radionuclide in the
femur. The 226Ra activity of the femurs was slightly reduced in the alginate-treated animals but the
amount of radium removed was independent of the injected dose that varied by a factor of 4. The
reduction in femur content presumably reflects decreased uptake due to reduced reabsorption of strontium
excreted into the gut, this was reflected in an increased faecal excretion in the treated animals.
A reduction in the absorption of 226Ra and 85Sr in mice following administration of aluminium
phosphate was reported by Kesley et al. [72K1].

9.6.3 Iodine
The substances recommended for human use are potassium iodide or iodate. These can be given orally in
the form of a suspension, or as Lugols solution which contains 50 mg iodine and 100 mg potassium
iodide per ml [80N1, 84W1, 92B1]. One blocking dose of 300 mg potassium iodide, if given within 30
min, will prevent further uptake by the thyroid. However, it may be advisable to administer 100 mg for a
further few days to prevent recycling of the radio-iodine. Potassium iodate, at similar doses, can be given
as an alternative to iodide. For current guidelines on iodine prophylaxis, and reviews of treatment
efficacy, the reader is referred to three recent publications [99H1, 99W2, 00G1].
9.6.3.1 Human data
One of the best examples of the efficacy of prompt and delayed treatment has used human volunteers
[67R1]. This work showed that if iodide administration is delayed by 6 h, the thyroid uptake is blocked
by only about 50 %, is whilst after 12 h, the uptake of iodine by the thyroid is scarcely affected by the
treatment.
If stable iodide is given after the first 24 h, there may be a prolonged retention of radio-iodine by the
thyroid due to the suppression of thyroid hormone release. Further, besides diluting the radio-iodine,
treatment with stable iodide and the massive increase in the iodine pool in the body also inhibits thyroid
metabolism. Under treatment with 300 mg sodium iodide followed a few daily doses of 100 mg, toxic
reactions are rare, although a few individuals may be over sensitive to iodide and develop angioedema. If
a reaction occurs, symptoms should disappear within a few days after cessation of treatment. Iodide
should also be administered with caution to persons with goitre or being treated for hyperthyroidosis
because the condition may exacerbate to thyrotoxicosis. This condition may also result if individuals have
a low dietary intake of iodine. Some people are allergic to large doses of iodide and such cases should be
treated with perchlorate.

9.6.4 Caesium
The substance recommended for the decorporation of caesium isotopes is Prussian Blue [80N11, 84W1,
92B1].
Landolt-Brnstein
New Series VIII/4

Ref. p. 9-31]

9 Decorporation of radionuclides

9-17

9.6.4.1 Human data


One of the major and most comprehensively investigated accidents involving internal contamination with
137
Cs occurred in Goiania in 1987 [94M1, 98I1]. Prussian Blue was administered orally to 46 individuals.
The dosages administered ranged from 1 to 3 g d1 for children, and from 3 to 10 g d1 for adolescents and
adults. In general treatment commenced about 10 d after exposure and continued over a period of about 3
weeks for children and over periods ranging from 3 weeks to 3 months for adults. During the
administration of the chelate, the mean retention half-times of 137Cs in the body were, on average, 43 %,
45 % and 69 % respectively of the values after termination of treatment. The committed effective doses
were reduced by between 1.7 fold and 6.2 fold, with a median value of 2.1 fold [94 M1, 98I1]. These
results are summarised in Table 9.4. The reduction in dose appeared to be independent of the dosage of
Prussian Blue, and the age of the patient [94M1, 98I1]. Reductions in doses of 2 to 3 fold have also been
found after other accidental intakes involving caesium isotopes [96M1, 85M1, 88T1].
Table 9.4. Committed whole body doses for individuals treated with Prussian Blue in the Goiania
accident [94M1, 98I1, 00S2]
Dose(1)
Dose(2)
[mSv]
Subject
Sex
Age [y]
Weight [kg] [mSv]
Ratio(3)
2.0
360
180
17
5
F
1
1.8
220
120
20
6
F
2
1.8
210
120
26
7
M
3
2.0
90
46
23
8
M
4
1.7
240
140
25
8
M
5
1.7
250
140
27
10
M
6
2.0
350
180
31
13
M
7
1.8
1200
700
38
13
M
8
3.3
670
200
55
13
M
9
1.7
39
22
55
13
M
10
1.9
68
35
58
14
M
11
2.0
290
140
50
19
M
12
5.5
5000
910
66
23
M
13
4.0
3800
970
69
28
M
14
3.6
3100
850
66
29
F
15
3.7
1400
370
61
32
M
16
2.7
140
49
80
33
M
17
2.4
390
160
58
36
F
18
6.2
1900
300
63
41
M
19
4.0
800
200
73
43
M
20
4.8
220
46
64
46
M
21
1

)With Prussian Blue treatment, 2)Without Prussian Blue treatment, 3)Reduction in dose with treatment.

9.6.4.2 Animal data


Many studies on the efficacy of Prussian Blue have been undertaken in laboratory animals [94M1, 96M1,
98I1], however in view of the detailed human studies that have been published they are not described
here.
Landolt-Brnstein
New Series VIII/4

9-18

9 Decorporation of radionuclides

[Ref. p. 9-31

In the last decade or so, work on improving the decorporation of caesium has involved investigations
of other hexacyanoferrates [90D1, 93D1, 91N1]. However, at present they do not appear to be
significantly more advantageous than Prussian Blue and it is difficult to see how substantial
improvements can be made.

9.6.5 Plutonium and americium


The currently recommended substances for enhancing biologically soluble forms of plutonium and
americium are CaDTPA or ZnDTPA (see Section 9.3).
9.6.5.1 Human data
Well-documented studies on the treatment of plutonium and americium after inhalation and wound
contamination have been published in the scientific literature. Perhaps the most notable example on the
efficacy of treatment is that often referred to as the Hanford americium accident, in which an individual
sustained an intake of about 41 MBq 241Am by inhalation and wound contamination resulting from the
explosion of an ion-exchange column [83B1, 89B1].
Briefly, in the absence of treatment, the bone and liver deposits were each predicted to be about
18,500 kBq, resulting in life-threatening doses of 0.07 Gy d1 and 1 Gy d1 respectively [89B1]. The
minimum 241Am contents of these tissues after an initial intravenous adminstration of CaDTPA followed
by the protracted administration of ZnDTPA by the same route, were about 220 and less than 4 kBq about
2 years after the accident. The values then increased to about 350 and 20 kBq by the 10th year as the
frequency of treatment was reduced. In total 583 g of DTPA was administered. Importantly no toxic side
effects were observed. The individual died 11 years after the accident from a medical condition unrelated
to the accident. A summary of the tissue content and excretion of 241Am is given in Table 9.5.
Table 9.5. Summary of tissue content and excretion of 241Am [89B1]
Organ content [kBq]
Time
Skin
Lungs
Bone
Liver
Day 0
185,000
Day 3
26,000
960
480
1,400
Day 10
14,000
290
320
590
Day 60
5,500
74
250
150
1 year
1,300
74
230
150
2 years
740
55 1)
220
ND
5 years
196
ND
280
9.6 2)
7 years
190
ND
NM
17
10 years
110
ND
350
19
11 years
NM
AS
NM
23 (AS)

Cumulative excretion [kBq]


Urine
Faeces
4,800
0
5,000
4,700
22,000
6,800
31,000
7,000
33,000
7,000
34,000
7,000
34,000
7,000
5.4 3)
1.4 3)
3)
4.8
0.036 3)
NM
NM

) not detected at 3 years


) increase in liver content due to reduction in DTPA treatment
3
) values per year based on 1-2 assays per year
ND not detectable, NM not measured, AS measured in autopsy sample
2

A good example of the comparative efficacies of surgical treatment and the efficacy of DTPA after the
intake of 239Pu oxalate through a puncture wound in the hand has also been reported [74S1]. It was
estimated that the wound was contaminated with 525 kBq of 239Pu. Cleaning the wound immediately after
the accident removed 144 kBq Pu. Continual monitoring of the wound with a probe showed that by 15 d,
about 285 Bq remained. During this time 17 kBq had been removed with surgical dressings and 16 kBq
Landolt-Brnstein
New Series VIII/4

Ref. p. 9-31]

9 Decorporation of radionuclides

9-19

was excreted in urine as a consequence of repeated DTPA treatment. The monitoring data implied that
37-74 kBq had been transferred to systemic tissues. A wide excision of the wound area performed in
hospital 15 d after the accident removed a further 255 kBq of 239Pu from the hand. Subsequent
measurements with a wound probe indicated that about 67 kBq remained at the wound site. In all there
were five courses of chelation therapy which commenced 40 minutes after the accident and were spread
over a period of 163 d. It was estimated that this treatment caused the elimination of 21 kBq of Pu in
addition to the 0.7 kBq that would be expected in its absence. Whilst the overall efficacy of treatment
with DTPA cannot be quantified due to the uncertainty in the estimate of systemic content, a reduction of
21 kBq would, based on the current ICRP model for 239Pu [93I1], imply an averted dose of 10.5 Sv
[00P1].
The published information on the effectiveness of oral treatment for inhaled plutonium and americium
is sparse. In general, it appears to have been useful for treating accidental intakes by workers [60N1,
67L1, 77S1]. However, uncertainties in the chemical forms of these actinides, the amounts inhaled and
the delays between exposure and treatment make assessments of its potential efficacy difficult.
Importantly however, the DTPA appeared to be of low toxicity; in one case 249 g of the free acid were
administered over a period of 16 weeks without any apparent side effects [67L1].
There are of course many other examples of the treatment of humans after accidental exposure to
plutonium and americium, published in the scientific literature. In many of these cases the administration
was only partially effective [60N1, 69S1, 72J1, 73S2, 76O1, 77S1, 80P1, 80V1, 89C1, 94W1]. In part
this may have been due to uncertainty in the chemical form of intake or exposure pattern, the
implementation of treatment regimens which may not have been optimised, or simply that the chemical
form was not amenable to treatment. Animal studies when properly executed need not suffer these
disadvantages. Moreover, such studies may also be used to evaluate alternative methods of treatment or
new substances.
9.6.5.2 Animal data
9.6.5.2.1 Inhaled plutonium and americium nitrate
Most animal experiments have been conducted with plutonium nitrate. In these circumstances it is
important that the mass concentrations of plutonium in tissues at the site of entry simulate a realistic
accident scenario, say intakes corresponding to doses up to two orders of magnitude greater than the
annual limit. Otherwise, the experimental data may provide information which could prejudice the use of
the ligand or the mode of administration.
Animal studies have shown that the administration of DTPA as an aerosol, by injection or orally in
drinking water can substantially reduce the lung deposit and hence systemic deposit of plutonium and
americium.
Information on the efficacy of inhaled DTPA in the rat after the inhalation of 238Pu and 241Am nitrate
is given in Tables 9.6 and 9.7 respectively. The tables show that concentrations of the chelate well below
the usual human dosage removed nearly all the contamination from the lungs. The small amounts retained
in other body tissues probably resulted from absorption and deposition in systemic tissues before the
commencement of treatment. It is noteworthy that the inhalation of DTPA was almost as effective as
repeated injection of the substance. These results contrast sharply with those obtained in the rat after
intakes in which the mass concentrations in the lungs were about 100 times higher. Under these
conditions aerosol DTPA was completely ineffective [77Bal].
Other studies, with the hamster, have shown that plutonium and americium can also be nearquantitatively removed from the lungs (i.e 1-3 % of controls) when either aerosol DTPA (2mol kg1) or
the combined administration of aerosol (2mol kg1) and injected (30 mol kg1) was delayed for up to
11 days after exposure. However in these cases, the total body contents of plutonium and americium
could be up to 30 % and 54 % of controls respectively, reflecting the difficulty in removing systemic
deposits that had accumulated before the commencement of treatment [00S2].

Landolt-Brnstein
New Series VIII/4

9-20

9 Decorporation of radionuclides

[Ref. p. 9-31

After inhalation of plutonium as nitrate by rats and treatment by intravenous injection, the siderophore
analogue 3,4,3-LI(1,2-HOPO) is appreciably more effective than DTPA, particularly after repeated
administration (Table 9.8). Under similar conditions of exposure and treatment, the ligands were
considered similarly effective for americium (Table 9.9).
The oral, and intraperitoneal, administration of DTPA has also been shown to be an effective method
of treatment in rats after the inhalation of plutonium and americium as their nitrates (Tables 9.10 and
9.11). Importantly, a ZnDTPA concentration an order of magnitude higher than that reported here did not
result in any observed histopathological changes to the liver, kidneys or gastrointestinal tract. It is also
noteworthy that the higher ZnDTPA concentration did not improve its efficacy. The low toxicity of
DTPA after oral administration is consistent with the data obtained from human [67L1] and other animal
[80T1, 90T1] studies.
The intravenous infusion and repeated injection of DTPA after the inhalation of plutonium nitrate by
dogs [92G1] have also been shown to be an effective method of treatment (Table 9.12).
Tables 9.6 to 9.12, see pages 9-21 and 9-22
9.6.5.2.2 Inhaled tributyl phosphate
Studies on the efficacy of injected DTPA and 3,4,3-LI(1,2-HOPO) have been undertaken after inhalation
by the rat. The repeated intraperitoneal injection of CaDTPA proved to be an effective treatment regimen
(Table 9.13). In another study involving a higher lung deposit and a shorter period of treatment (Table
9.14), the reduction in the plutonium contents of the lungs and systemic tissues were less, but the study
emphasised the higher efficacy of 3,4,3-LI(1,2-HOPO).
Table 9.13. Efficacy of injected DTPA on
retention of 238Pu in rats after inhalation as TBP
[85S2, 00S2]
% controls at 28d
( SE, N=5)(1)
Treatment
Lungs
Total body
CaDTPA(2)
4.3 0.7 16 2
ZnDTPA(2)
2.5 0.6 15 2
4.2 0.8 26 2
ZnDTPA(3)
Initial lung deposit 384 Bq 238Pu, 0.59 ng Pu.
Equivalent intake of Pu-239 by workers 0.86104 ng or
1.98104 Bq, i.e. 32 ALIs (CED 640 mSv).
1
)% inhaled activity in controls at 1 d, lungs 23.1 1.8,
total body 62.0 3.0.
% inhaled activity in controls at 28 d, lungs 9.5 1.0,
total body 43.1 2.7.
2
)30 mol kg1 CaDTPA or 200 mol kg1 ZnDTPA
injected i.p. at 30 min, 6 h, 1 d, 2 d, 5 d, and then every
3-4 d to 26 d.
3
)Treatment regimen as for (a) but delayed for 1 d.

Table 9.14. Efficacy of injected 3,4,3-LI(1,2HOPO) and CaDTPA on retention of 238Pu in rats
after inhalation as TBP [93P1, 00S2]
% controls at 7d
( SE , N=4)
Treatment
Lungs
Skeleton
iv injection(1)
LIHOPO
27 2 12 3
CaDTPA
30 1 22 2
im + iv injection(2)
LIHOPO
28 2 2.9 0.9
CaDTPA
45 2 14 2
1

) Initial lung deposit 5200 400 Bq, 8.1 ng Pu.


Equivalent intake of Pu-239 by workers 1.18105 ng or
2.72105 Bq, i.e. 435 ALIs (CED 8.7 Sv)
Administration of 30 mol kg1 after 1 h
Liver contents were 6.0 0.7 % and 17.5 3.4 % of
those in controls after administration of LIHOPO and
DTPA respectively.
2
) Initial lung deposit 34000 3000 Bq, 53 ng Pu.
Equivalent intake of Pu-239 by workers 7.70105 ng or
1.78106 Bq, i.e. 2840 ALIs (56.8 Sv)
Administration of 30 mol kg1 after 1h (iv) and 1 d and
2 d (im).
Liver contents were 1.7 0.2 % and 8.1 2.2 %
respectively of those in controls after administration of
LIHOPO and DTPA respectively.

Landolt-Brnstein
New Series VIII/4

Ref. p. 9-31]

9 Decorporation of radionuclides

9-21

Table 9.6. Efficacy of aerosol and injected DTPA


on retention of 238Pu in rats after inhalation as
nitrate [85S1, 00S2]. = arithmetic mean, N =
number of observations.
% controls at 28 d
( SE, N=5)(1)

Table 9.7. Efficacy of aerosol and injected DTPA


on retention of 241Am in rats after inhalation as
nitrate [85S1, 00S2]
% controls at 28d
( SE, N=5)(1)
(2)
Treatment
Lungs
Total body

Treatment(2)
Aerosol(3)
Aerosol plus
injection(4)
Injection only(5)

Aerosol(3)
2.3 0.5
(4)
Aerosol plus injection
1.6 0.3
5.0 2.1
Injection only(5)

Lungs
2.1 1.1

Total body
7.6 1.2

1.1 0.1
4.4 2.4

4.2 0.7
5.7 1.4

Initial lung deposit, 505 37 Bq, 0.78 ng Pu.


Equivalent intake of 239Pu by workers 1.13104 ng or
2.61104 Bq, i.e. 42 ALIs (CED 840 mSv).
1
) % ILD in controls at 28 d; lungs, 29.3 3.8, total
body, 45.1 5.8.
2
) DTPA administration at 30 min, 6 h, 1 d, 2 d, 3 d, 5 d
and then twice weekly to 27 d; first administration
CaDTPA, then ZnDTPA.
3
) Inhalation, 2 mol kg1.
4
) Inhalation, 2 mol kg1and intraperitoneally (i.p.)
injection, 30 mol kg1
5
) i.p. injection, 30 mol kg1

Table 9.8. Efficacy of injected 3,4,3-LI(1,2LIHOPO) and DTPA on retention of 238Pu in rats
after inhalation as nitrate [92S1, 00S2]
% controls at 7d
( SE, N=4)(1)
(2)
Treatment
Lungs
Total body
LIHOPO(3)
DTPA

(3)

LIHOPO
DTPA

(4)

(4)

LIHOPO(5)

11 1

11 1

16 2

18 2

1.8 0.3

11 1

Landolt-Brnstein
New Series VIII/4

Initial lung deposit, 350 25 Bq, 2.8 ng Am.


Equivalent intake of 241Am by workers 4.05104 ng or
5.09106 Bq, i.e. 6870 ALIs (CED 137 Sv).
1
) % ILD in controls at 28 d; lungs, 14.3 1.8, total
body 30.9 4.1.
2-5
) Treatment regimens as given in Table 9.7

Table 9.9. Efficacy of injected 3,4,3-LI(1.2HOPO) and DTPA on retention of 241Am in rats
after inhalation as nitrate [92S1, 00S2]
% controls at 7d
( SE, N=5)(1)
(2)
Treatment
Lungs
Total body
LIHOPO(3)
41 4
31 3
DTPA(3)

21 2

15 1

LIHOPO (4)

13 2

11 2

13 2

91

81 8

64 4

12 1

4.5 0.4

DTPA (4)

24 2

27 2

LIHOPO

Initial lung deposit, 600 25 Bq, 0.92 ng Pu.


Equivalent intake of 239Pu by workers 1.34104 ng or
3.08104 Bq i.e. 49 ALIs (CED 980 mSv).
1
) % ILD in controls at 7 d: lungs, 64.6 4.4, total body
86.3 4.8.
2
) DTPA, 30 mol kg1 administered by
intraperitoneally (i.p.) injection; first administrations
CaDTPA then ZnDTPA .
3
) 30 min only.
4
) 30 min, 6 h, 1 d ,2 d ,3 d.
5
) 1d only.

3.7 0.6
2.9 0.5
3.6 0.9

(5)

Initial lung deposit, 623 25 Bq, 4.97 ng. Am.


Equivalent intake of 241Am by workers 7.22104 ng or
9.05106 Bq, i.e. 12,230 ALIs (CED 244 Sv).
1
) % ILD in controls at 7 d : lungs, 40.4 3.7, total
body, 71.5 4.4.
2-5
) Treatment regimen as in Table 9.8

9-22

9 Decorporation of radionuclides

[Ref. p. 9-31

Table 9.10. Efficacy of oral and intraperitoneally


(i.p.) injected ZnDTPA on retention of 238Pu and
241
Am in rats after inhalation as nitrate: prompt
administration [95G1, 00S2]
% controls at 21 d
( SE, N=4)(1)
(2)
Treatment
Lungs
Total body
Plutonium
Oral(3)
2.2 0.4 8.8 1.5
Oral(4)
2.2 0.3
7.8 0.8
1.7 0.3 5.2 0.7
i.p(5)
Americium
Oral(3)
3.6 0.6 6.0 0.6
3.2 0.3 4.8 0.5
Oral(4)
i.p(5)
1.7 0.6 2.5 0.4

Table 9.11. Efficacy of oral and intraperitoneally


(i.p.) injected ZnDTPA on retention of 238Pu and
241
Am in rats after inhalation as nitrate: delayed
administration [93S1, 00S2]
% controls at 28 d
( SE , N=4)(1)
(2)
Treatment
Lungs
Total body
Plutonium
Oral(3)
6.1 0.4 19 3
Oral(4)
6.2 0.3 17 2
11 1
25 4
i.p(5)
Americium
Oral(3)
3.6 0.6 23 3
3.2 0.3 20 3
Oral(4)
i.p(5)
1.7 0.6 29 4

Initial lung deposit, Pu 676 96 Bq, 1.04 ng Pu , Am,


354 49 Bq , 2.82 ng Am.
Equivalent intake by workers: 239Pu 1.51104 ng or
3.49104 Bq, i.e. 56 ALIs (CED 1120 mSv); 241Am
4.10104 ng or 5.14106 Bq, i.e. 6950 ALIs (CED
139 Sv).
1
) % ILD in controls at 21 d: Pu, lungs, 41.0 3.6 , total
body 63.9 4.7;
Am, lungs, 20.1 1.6, total body 51.7 4.
2
) Treatment commenced 1 h after exposure.
3
) 950 mol kg1 d1 for 21d.
4
) 95 mol kg1 d1 for 21d.
5
) 30 mol kg1 twice weekly for 21 d.

Initial lung deposit, Pu 676 96 Bq, 1.04 ng Pu , Am,


354 49 Bq , 2.82 ng Am.
Equivalent intake by workers: 239Pu 1.51104 ng or
3.49104 Bq, i.e. 56 ALIs (CED 1120 mSv); 241Am
4.10104 ng or 5.14106 Bq, i.e. 6950 ALIs (CED
139 Sv)
1
) % ILD in controls at 28 d: Pu, lungs, 30.7 2.7, total
body 60.0 4.9; Am, lungs, 14.8 1.0, total body 51.3
3.8.
2
) Treatment commenced 7 d after exposure.
3-5
) Treatment as given in Table 9.10.

Table 9.12. Efficacy of injected CaDTPA on


retention of 238Pu in dogs after inhalation as
nitrate [93G1, 00S2]
% controls at 64d
( SD, N=2)(1)
Treatment
Lungs
Total body
DTPA injections(2,3)
20 6
22 4
DTPA infusions(4,5)
22 6
17 2

) CaDTPA i.v. (30 mol. kg.-1 ) after 1 h, 1 d , 2 d,


3 d, 4 d, and ZnDTPA twice weekly thereafter.
3
) Liver and bone content reduced to 8.3 4.2 % and
36 5 % of controls.
4
) CaDTPA i.v. after 1 h ,then subcutaneous infusion
with ZnDTPA (30 mol kg1 d1 ) from 1 d.
5
) Liver and bone content reduced to 3.8 1.4 % and
28 6 % of controls.

) Initial lung deposit, 16-26 kBq.


% initial deposit in controls after 64 d , lungs 10.8
2.9, liver 31.3 4.6 , bone 30.5 3.3 total body
76 4.

9.6.5.2.3 Inhaled plutonium dioxide


Under normal conditions the soluble or ultrafine component of 239Pu dioxide aerosols would be expected
to be appreciably less than 1 % [72ICRP]. Hence the administration of DTPA by whatever route would
have little impact on reducing the committed effective dose.
However the presence of other metals during the formation of the aerosol, particularly those of low
atomic weight such as sodium, can substantially increase the ultrafine component, and in such
circumstances the administration of the ligand will be much more effective [80S1]. The experimental data
are summarised in Table 9.15.
Landolt-Brnstein
New Series VIII/4

Ref. p. 9-31]

9 Decorporation of radionuclides

9-23

The absorption of 238Pu into blood after the inhalation of 238PuO2 is governed by the formation of
particles about 1 nm in diameter by radiolytic fragmentation. The transportable fraction arising from this
process is retained in part in the lungs from where it can be mobilised by DTPA [82S1]. The experimental
data summarised in Table 9.16 illustrate that DTPA was effective for removing Pu from the lungs as
judged by the appreciable increase in urinary excretion. On the other hand the reduction in the body
content after treatment was only about 20 % as a consequence of the competing action of mucociliary
clearance. It is concluded that the protracted treatment required for a small reduction in the lung content
would be unlikely to be used in humans, where for large intakes lung lavage would be more beneficial.
Table 9.15. Efficacy of injected CaDTPA on retention of Pu and Am in hamsters after inhalation of a
mixed aerosol of PuO2 (+AmO2) and Na2O, Pu:Na atomic ratio 1:30 [80S1]
% controls at 30 d ( SE, N= 8) (1)
Treatment (2)

Lungs

Total body

Pu

33 6

22 3 (c)

Am

40 7

20 3 (d)

) Initial deposit, 3.6 kBq kg1 body mass;


% ILD in controls at 30 d; Pu: lungs 27.4 4.6, total body 81.2 8.2, Am: lungs 21.0 3.0, total body 83.0 8.5
2
) DTPA, 30 mol kg1 andimistered i.p. at 3 h, 1 d, 2 d, 4 d.
3
) % controls in liver and bone, 13 3 and 17 4 respectively
4
) % contols in liver and bone, 11 2 and 16 3 respectively
1

Table 9.16. Efficacy of DTPA on retention and excretion of 238Pu in the hamster after inhalation as
238
PuO2 [82S1]
Treatment
% body deposit at 7 d ( SE, N=4 - 6) (1)
Lungs
Systemic Urine
Faeces
% ILD at 154 d
aerosol DTPA (2)
31.5 2.7
4.0 0.2 23.5 1.0
41.0 3.6
controls
39.2 1.1
3.1 0.4
2.8 0.2
54.9 1.4
% ILD at 147 d
aerosol (2)+ i.p (3) DTPA
30.8 1.4
4.1 0.1 29.6 0.7
35.5 2.4
controls
37.7 1.1
7.6 0.6
5.2 0.5
49.5 0.8
1

) Initial body content of 238PuO2 at 7 d, 510 40 Bq, of which 98.8 % was in the lungs.
) DTPA administration, 2 mol kg1 commenced 7 d after exposure and continued at weekly intervals to 147 d
3
) DTPA injections, 26 mol kg1, commenced 10 d after exposure and continued at weekly intervals until 143 d
2

9.6.5.2.4 Inhaled americium dioxide


The efficacy of DTPA for inhaled 241AmO2 has been investigated after administration as an aerosol and
intraperitoneal injection in the hamster [84S1] and by intravenous injection and infusion using implanted
osmotic pumps in the dog [88G1]. All methods of administration were moderately effective (Tables 9.17,
9.18). The latter treatment was particularly impressive since it virtually prevented the deposition of 241Am
in systemic tissues. Implanted osmotic pumps in humans for long periods may be impracticable. However
the data obtained for 241Am nitrate after the oral administration of DTPA referred to above suggest that
this could be an alternative mode of administration for inhibiting systemic deposition.

Landolt-Brnstein
New Series VIII/4

9-24

9 Decorporation of radionuclides

Table 9.17. Efficacy of injected ZnDTPA for


241
Am nitrate and 241AmO2 inhaled by the hamster
[84S1, 00S2]
% controls at 74 d
( SE, N=5)
Treatment
Lungs
Total body
Am nitrate(1)
Injection(3)
31
13 1
Inhalation(4)
31
54 4
Am dioxide(2)
Injection(3)
14 2
16 2
Inhalation(4)
27 4
56 4
ILD 150 Bq or 80 Bq at the commencement of
treatment.
1
) % ILD in controls at 74 d, lungs, 19.8 2.6, total
body, 54.4 2.6.
2
) % ILD in controls at 74 d, lungs, 25.2 2.4, total
body, 58.6 3.1.
3
) Zn DTPA injected intaperitoneally at weekly intervals
from 4 d to 67 d at a dosage of 200 mol kg1.
4
) Zn DTPA inhaled at weekly intervals from 4 d to 67 d
at a dosage of 2 mol kg1.

[Ref. p. 9-31

Table 9.18. Efficacy of CaDTPA on retention of


Am in dogs after inhalation of AmO2 [88G1,
00S2]
% controls at 64 d
( SD, N=2)1
Treatment
Lungs
Total body
DTPA
injections(2,3,5)
63 25 29 10
DTPA
30 9
11 3
infusions(4,5)

241

) Initial lung deposit, 17-39 kBq.


% initial deposit in controls after 64 d, lungs 24.5 2.5,
liver 25.1 3.4, bone 21.8 4.2, total body 76.7 7.8.
2
) CaDTPA i.v. (30 mol kg1 ) after 1 h, 1 d, 2 d, 3 d,
4 d, and ZnDTPA twice weekly thereafter.
3
) Liver and bone content reduced to 4.7 3.2 % and
18 4 % of controls.
4
) CaDTPA i.v. after 1 h ,then subcutaneous infusion
with ZnDTPA (30 mol kg1 d1 ) from 1 d.
5
) Liver and bone content reduced to 0.43 0.28 % and
1.7 0.7 % of controls.

9.6.5.2.5 Wound contamination with plutonium and americium nitrate


For the purpose of investigating new or alternative treatment protocols, human wounds are usually
simulated in animals by subcutaneous or intramuscular injection of radionuclides. The ligands have been
administered as either a single and repeated local administration or combinations of local and
intraperitoneal injections, intravenous injection and oral administration. In general, local administration
has proved to be most effective and intravenous injection ineffective.
The comparative efficacies of 3,4,3-LI(1,2-HOPO) and DTPA in rats after the intramuscular injection
of plutonium and americium nitrate are shown in Tables 9.19 and 9.20. The data show that virtually all of
the plutonium and americium were removed from the body by a single local injection of 30 mol kg1 of
3,4,3-LI(1,2-HOPO). The retention of plutonium and americium in the body using a similar treatment
protocol with DTPA were about 30 and 20 times more respectively.
The efficacy of both ligands falls appreciably with delayed administration. However the retention of
plutonium and americium at the wound site and in the total body is still about 3 to 4 times less with
3,4,3-LI(1,2-HOPO) than with DTPA (Tables 9.21, 9.22).
The high efficacy of 3,4,3-LI(1,2-HOPO) after wound contamination has also been reported elsewhere
[96V1].

Landolt-Brnstein
New Series VIII/4

Ref. p. 9-31]

9 Decorporation of radionuclides

9-25

Table 9.19. Efficacy of injected 3,4,3-LI(1,2HOPO) and DTPA on retention of 238Pu after
intramuscular injection as nitrate: prompt
treatment [93S1, 00S2]
% controls at 7 d
( SE, N=4)(1)
Treatment
Wound site Total body
LIHOPO(2)
4.8 0.4
5.9 0.5
LIHOPO(3)
0.9 0.1
0.9 0.1
0.6 0.1
1.0 0.1
LIHOPO(4)
DTPA(5)
33 2
32 1
LIHOPO (6)
33 3
33 2

Table 9.20. Efficacy of injected 3,4,3-LI(1,2HOPO) and DTPA on retention of 241Am after
intramuscular injection: prompt treatment [93S1,
00S2]
% controls at 7 d
( SE, N=4)(1)
Treatment
Wound site Total body
LIHOPO(2)
8.5 0.1
8.8 0.6
LIHOPO(3)
0.6 0.1
0.8 0.1
0.4 0.1
1.2 0.1
LIHOPO (4)
DTPA(5)
27 3
22 2
LIHOPO(6)
43 4
39 2

) Injected activity 190 5 Bq , 0.3 ng Pu.


% injected activity in controls at 7 d, wound site
70.2 1.7 total body 95.7 1.0.
2
) 3 mol kg1 locally at 30 min.
3
) 30 mol kg1 locally at 30 min.
4
) 30 mol kg1 at 30 min, plus i.p. at 6 h, 1 d, 2 d and
3 d.
5
) as (4) with CaDTPA for local injection and ZnDTPA
for i.p.
6
) 30 mol kg1 i.v. at 30 min.

Table 9.21. Efficacy of injected 3,4,3-LI(1,2HOPO) and DTPA on retention of 238Pu after
intramuscular injection: delayed treatment [94G1,
00S2]
% controls at 7 d
( SE, N=4)(1)
Treatment
Wound site Total body
LIHOPO (2)
1.0 0.1
1.2 0.1
CaDTPA(2)
39 2
31 2
17 1
15 1
LIHOPO (3)
Ca DTPA (3)
71 2
76 2
LIHOPO (4)
24 1
23 1
75 2
81 2
CaDTPA(4)

Table 9.22. Efficacy of injected 3,4,3-LI(1,2HOPO) and DTPA on retention of 241Am after
intramuscular injection: prompt and delayed
treatment [94G1, 00S2]
% controls at 7 d
( SE, N=4)(1)
Treatment
Wound site Total body
LIHOPO (2)
0.7 0.1
1.0 0.1
CaDTPA(2)
28 2
25 2
15 1
13 1
LIHOPO(3)
CaDTPA(3)
69 2
68 2
LIHOPO(4)
23 1
22 1
72 2
73 2
CaDTPA(4)

) Injected activity 200 Bq , 0.3 ng Pu.


% injected activity in controls at 7 d, wound site
68.4 0.9, total body 97.2 1.3.
2
) 30 mol kg1 locally at 30 min.
3)
30 mol kg1 locally at 6 h
4)
30 mol kg1 at 1 d.

) Injected activity 200 5 Bq , 1.6 ng Am.


% injected activity in controls at 7 d, wound site
70.5 2.6 total body 96.8 2.0.
2-6
) Treatment protocols as given in Table 9.19

) Injected activity 200 Bq, 1.6 ng Am.


% injected activity in controls at 7 d, wound site
71.4 1.1, total body 97.0 1.0.
2-4
) Treatment protocols as given in Table 9.21.

9.6.5.2.6 Wound contamination with plutonium tributylphosphate (TBP)


The protocols used have in broad terms been similar to those used for plutonium nitrate. A summary of
the data obtained after the intramuscular injection of plutonium-TBP is given in Table 9.23. They show
that the removal of plutonium from the wound site and systemic tissues is considerably less than for
plutonium nitrate. This is attributed in part to the greater mass of plutonium deposited, but clearly the
influence of chemical form is also important. It is noteworthy that the skeletal and liver contents after the
administration of 3,4,3-LI(1,2-HOPO) are 3 to 4 times less than with DTPA.

Landolt-Brnstein
New Series VIII/4

9-26

9 Decorporation of radionuclides

Table 9.23 Efficacy of injected 3,4,3-LI(1,2HOPO) and DTPA on retention of 238Pu after
intramuscular injection as TBP [95P1, 00S2]
% controls at 7 d
( SD, N=6)(1)
Treatment
Wound site Total body
Local injection
LIHOPO(2)
73 24
60 20
CaDTPA(2)
71 15
72 15
Iv injection
86 13
70 13
LIHOPO(2)
CaDTPA(2)
100 22
87 15

[Ref. p. 9-31

) % injected activity in controls at 7 d, wound site


71.1 9.8 , total body 97.6 8.9.
2
) 30 mol kg1 CaDTPA after 30 min.

9.6.6 Thorium
The recommended substance for thorium isotopes is DTPA (see Section 9.3). The authors are unaware of
any definitive human data which demonstrates the efficacy of the substance.
9.6.6.1 Animal data
9.6.6.1.1 Inhalation of thorium nitrate
Studies in rats [91S1] have shown that DTPA is poorly effective when the amounts of 232Th simulated
acute intakes equivalent to the dose limit, even when the substance was injected at dosages of 300 and
1000 mol kg1 (Table 9.24). It is noteworthy that treatment is unlikely to be implemented for intakes of
less than 10 times the dose limit (see Section 9.2). Compared with the human equivalent dosage of
DTPA, the increase in efficacy for 232Th using 3,4,3-LI(1,2-HOPO) could only be considered marginal
(Table 9.25).
The efficacy of DTPA increased to a moderate extent when substantially lower mass concentrations of
thorium were used, as with 228Th. (Table 9.26). However the most effective treatment to date has involved
the repeated injection of 3,4,3-LI(1,2-HOPO) whereby the thorium content of the lungs was reduced to
17 % of that in controls (Table 9.24). The results obtained with 3,4,3-LI(1,2-HOPO) suggest that the
ineffectiveness of DTPA is unlikely to be due to the formation of hydrolysis products in the lungs.
9.6.6.1.2 Wound contamination with thorium nitrate
Simulated wound studies in rats have been undertaken mainly with the high specific activity 228Th
isotope. The data given in Table 9.27 show that 7 to 8-fold reductions in the body content occur after the
prompt local injection of 3,4,3-LI(1,2-HOPO) followed by repeated administration at the same dosage.
Under the same conditions, the reduction after DTPA administration was about 2-fold. The table also
shows that the efficacy of both ligands is reduced appreciably when treatment is delayed only by 1 day.
Based on other data with plutonium and americium [00S2], it is unlikely that intravenous or intraperitonal
injections alone would have any beneficial effect.

Landolt-Brnstein
New Series VIII/4

Ref. p. 9-31]

9 Decorporation of radionuclides

Table 9.24 Efficacy of single and repeated DTPA


administration on retention of 232Th in rats after
inhalation as nitrate (1) [91S1]
% controls at 7 d
( SE, N=4)(2)
Treatment (3)
CaDTPA, (4)
CaDTPA(5)
Ca+ ZnDTPA(6)
CaDTPA (7)

Lungs
75 8
66 7
66 9
98 10

Total body
74 8
65 8
65 8
97 10

) initial lung deposit of 230+232Th, 586 16 Bq;


6.46 0.18 g Th
2
) % of ILD in control animals at 7 d: lungs
76.2 5.2, total body 82.0 5.4
3
) chelates administered intraperitoneally
4
) 300 mol kg1 administered at 30 min only
5
) 1000 mol kg1 administered at 30 min only
6
) 1000 mol kg1 CaDTPA administered at 30 min
and 300 mol kg1 ZnDTPA at 1 d, 2 d and 3 d
7
) 1000 mol kg1 administered at 1 d only

Table 9.26 Efficacy of injected 3,4,3-LI(1,2HOPO) and DTPA on retention of 228Th in rats
after inhalation as nitrate [98S1, 00S2]
% controls at 7d
( SD, N=4)(1)
Treatment
Lungs
Total body
LIHOPO(2)
36 3
29 2
LIHOPO(3)
17 2
17 2
73 4
78 3
DTPA(3)
1

) Initial lung deposit, 4 ng Th


Equivalent intake of Th by workers, 58 mg or 4.18104
Bq 230Th and 1.74109 Bq 228Th i.e. 59 ALIs 230Th
(CED 100 mSv) and 2106 ALIs 228Th (2000 Sv)
% ILD in controls at 7 d: lungs 50.7 1.9, total body
69.9 3.5.
2
) 30 mol kg1 i.p. after 30 min
3
) 30 mol kg1 i.p. after 30 min, 6 h, 1 d, 2 d, 3 d

9-27

Table 9.25 Efficacy of injected 3,4,3-LI(1,2HOPO) and DTPA on retention of 232Th in rats
after inhalation as nitrate(1) [91S1, 98S2]
% controls at 7 d
Treatment
( SD, N=4)(1)
Lungs
Total body
LIHOPO (2)
93 7
87 5
LIHOPO (3)
73 6
69 5
ZnDTPA (3)
93 6
91 5
1

) Initial lung deposit, 4.2 g Th


Equivalent intake of Th by workers 61 mg or 247 Bq
232
Th, i.e. 0.36 ALI for 232Th (CED 7.2 mSv)
% ILD in controls at 7 d: lungs 69.9 4.5, total body
78.1 4.6
2
) 30 mol kg1 i.p. after 30 min
3
) 30 mol kg1 i.p. after 30 min, 6 h, 1 d, 2 d and 3 d

Table 9.27 Efficacy of injected 3,4,3-LI(1,2HOPO) and DTPA on retention of 228Th after
intramuscular injection: prompt and delayed
treatment [95S1, 00S2]
% controls at 7 d
( SE, N=4)(1)
Treatment
Wound site
Total body
LIHOPO (2)
14 1
20 1
CaDTPA(2)
60 3
65 3
LIHOPO(3)
12 1
15 1
Ca DTPA(3)
50 3
55 2
LIHOPO(4)
38 2
40 2
CaDTPA(4)
79 3
79 2
1
) Injected activity 600 Bq 228Th, 0.1 ng Th.
% injected activity in controls at 7 d, wound site 64.8
1.5, total body 91.5 1.7.
2
) 30 mol kg1 locally at 30 min
3
) 30 mol kg1 locally at 30 min and then by ip
injection at 6 h, 1 d, 2d and 3d after exposure.
4
) 30 mol kg1 at 1 d and then by i.p. injection at 6 h,
1 d, 2 d and 3 d later.

9.6.7 Uranium
At the present time no agent can be recommended for the removal of uranium from the systemic
circulation. Uranium complexation by sodium bicarbonate has been proposed for reducing the systemic
deposit [80N1, 84W1, 92B1]. However, this is not supported by controlled studies with laboratory
animals under realistic conditions e.g. with delays between exposure and treatment of 30 min or more.
Landolt-Brnstein
New Series VIII/4

9-28

9 Decorporation of radionuclides

[Ref. p. 9-31

The recommended dose is 250 cm3 of 1.4 % sodium bicarbonate (42 mmol) administered by slow
intravenous infusion, with further infusions on subsequent days if necessary [92B1]. Since the plasma
bicarbonate concentration is held more or less constant at ~25 mmol dm3, and its turnover time is fairly
rapid, it appears unlikely that the slow infusion of a further 42 mmol would lead to a sufficiently large
and sustained increase in plasma bicarbonate concentration to significantly enhance the excretion of
uranium. It should also be noted that alkalosis, respiratory acidosis and hypokalaemia may result from
such treatment.
The authors are unaware of any definitive human data which demonstrates the efficacy of bicarbonate,
or indeed any other substance.
9.6.7.1 Animal data
9.6.7.1.1 Injection of uranium
Other than the octoxide and dioxide, uranium compounds formed in the nuclear fuel cycle are readily
absorbed into the blood after inhalation e.g. ammonium diuranate, uranyl nitrate, uranium tetrafluoride.
Since the role of treatment will be to minimise nephrotoxocity in the early lung clearance phase, the
administration of uranium by intravenous injection would be appropriate for evaluating the likely efficacy
of various treatment regimens.
Since there appears to be no substantive evidence that the administration of sodium bicarbonate is an
effective method of treatment, several alternative substances have been investigated in animals. Some of
these such as tiron, certain phosphonates and hydroxypyridonate derivatives have caused large reductions
in the kidney and skeletal contents of uranium when administered simultaneously or within minutes
[00S2, 00S3]. However under more realistic conditions when delays between exposure and treatment may
be 30 minutes, or probably longer, they are poorly effective. The efficacies after immediate and delayed
treatment for some selected substances are shown in Tables 9.28 and 9.29. Notably, Table 9.29 also
demonstrates the poor efficacy achieved with sodium bicarbonate.
9.6.7.1.2 Wound contamination
The ligand 3,4,3-LI(1,2-HOPO) has been shown to be the most effective yet tested for plutonium,
americium and thorium after inhalation and wound contamination. However the data in Table 9.29 show
that it is only moderately effective even when administered immediately after the uranium. Under more
realistic conditions of exposure and treatment it is poorly effective. The immediate administration of the
bisphosphonate EHBP, (ethane-1-hydroxy-1,1-bisphosphonate) has been shown to prevent death in
animals [94U1, 00M1]. However, the prevention of death in an acutely poisoned animal is not a reliable
indicator of the effect of a chelator at the much lower level of human contamination that would be
expected in any likely industrial accident, further the doses of EHBP used were more than 100 times
those used in the treatment of human disease. The data given in Table 9.30 demonstrates that the efficacy
of EHBP again falls rapidly with time. The main interest in EHBP is that some preparations have been
licensed for other medical purposes, and hence its toxicity and metabolism are well known.
It is concluded that at present no effective substance is available for the treatment of internal
contamination by uranium.

Landolt-Brnstein
New Series VIII/4

Ref. p. 9-31]

9 Decorporation of radionuclides

Table 9.28 Efficacy of injected polyphosphonic


acids on retention of uranium [92G1, 98H1, 00S2]
% controls at 4 d
( SE, N=4)(1)
Treatment
Kidneys
Total body
Immediate
HMDTMP(2)
81
31 3
DTPMP(2)
81
32 3
Delayed 30 min
HMDTMP(2)
60 8
81 6
DTPMP(2)
70 9
86 6
1
) ID 300 Bq 233U, Ligand:uranium mol ratio 1.5104
% ID in controls, kidneys, 9.90.9, total body, 27.31.3
2
) 300 mol kg1 of hexamethylenediaminetetrameyhylene-phosphonic acid or diethylenetriaminepentamethylene-phosphonic acid.

9-29

Table 9.29 Efficacy of injected 3,4,3-LI(1,2HOPO) and sodium bicarbonate on retention of


uranium [95H1, 98 H1, 00S2]
% controls at 24 h
( SE, N=5)(1)
Femora
Treatment
Kidneys
(Bone)
Uranium im
Chelate im(2)
LIHOPO
23 3
46 5
NaHCO3
87 5
67 22
Uranium im,
chelate ip(3)
LIHOPO
54 9
82 13
NaHCO3
64 13
114 33
Uranium iv,
chelate iv(4)
LIHOPO
21 4
61 8
NaHCO3
76 20
102 25
) Initial deposit, 0.84 mol kg1.
) Treatment immediate. % ID in controls, kidneys 18.2,
femora 1.4.
3
) Treatment after 30 min. % ID in controls, kidneys
15.5, femora 1.25.
4
) Treatment immediate. % ID in controls, 15.3, femora
2.2. LIHOPO, 30 mol kg1, NaHCO3, 640 mol kg1 in
all cases.
1
2

Table 9.30 Removal of intramuscular 233U from


rats by intramuscular injection of EHBP [98H1,
00S2]
% controls at 24 h(1)
EHBP
Kidneys
Total body
5 min
24 7
70 9
30 min
55 21
89 12

) Mean standard error, n=5


U injected 0.02 mol kg1, 1.5 kBq kg1, ligand to U
molar ratio, 5000.
% initial deposit in controls: kidneys, 14.1 2.9, total
body 54.6 6.0
EHBP = ethane-hydroxy-1,1-bisphosphonate

9.7 Future research needs


For insoluble substances deposited in the lungs or in wounds, decorporation procedures involving lung
lavage and surgical excision respectively are the most appropriate. Increasing the expertise in the
treatment of obstructive lung disease should increase the availability of the lavage procedure after major
accidents. However, it is difficult to perceive what improvements could be made to increase the efficacy
of lavage, and surgical excision, other than through good medical practice and medical-team training.
Likewise, it is difficult to envisage what improvements can be made to increase the elimination of
tritium and iodine isotopes from the body beyond those procedures described earlier.

Landolt-Brnstein
New Series VIII/4

9-30

9 Decorporation of radionuclides

[Ref. p. 9-31

The decorporation of caesium becomes an important issue in major radiation accidents e.g.
contamination of members of the public from leaking radiotherapy sources, fission product release in a
nuclear accident. The commonly used decorporating agent, Prussian Blue, is of only limited efficacy and
it is recommended therefore that more effective agents are developed and tested.
Within the last decade in particular, considerable progress has been made in evaluating treatment
regimens and developing new chelating agents for the decorporation of plutonium, americium, and high
specific activity forms of thorium. Animal studies have shown that some of these ligands are much
superior to DTPA, comprehensive toxicity testing has not yet been undertaken, and for practical purposes
DTPA must at present remain the agent of choice for these elements. It is essential, however, that patients
who receive DTPA therapy are followed up in order to establish the efficacy of the treatment regimen for
the particular physico-chemical form.
Little progress has been made on the decorporation of uranium. In view of the large scale and
widespread use of the element, this should be viewed with some concern. It is recommended that research
to find new methods for the decorporation of uranium should be expedited.

Landolt-Brnstein
New Series VIII/4

9 Decorporation of radionuclides

9-31

9.8 References
60N1
64W1
65H1
67H1
67L1
67R1
67S1
67S2
68C1
68H1
69N1
68S1
68S2
68V1
68W1
69S1
69S2
71L1
71V1
71V2
72H1
72H2
72H3
72I1
72J1
72K1
72V1
73S1
74S2
73S3
76O1
77B1
77S1
77V1

Norward, W.D., in: Proceedings of the 13th International Congress on Occupational Health,
New York, 1960, p. 335.
Waldron-Edward, D., Paul, T.M., Skoryna, S.C.: Can. Med. Assn. J. 91 (1964) 1006.
Hesp, R., Ramsbottom, B.: PG Report 686 UKAEA, 1965.
Hesp, R., Ramsbottom, B.: Strontium Metabolism (Leniham, J.M.A., Loutit, J.M., Martin,
J.H. eds.) Academic Press, London (1967) 311.
Lagerquist, C.R., Putzier, C.A., Piltingsgrud, C.W.: Health Phys. 13 (1967) 965.
Ramsden, D.,Passant, F.H.,Peabody, C.O.,Speight, R.G.: Health Phys. 13 (1967) 633.
Spencer, H., Lewin, I., Samachson, J.: Int. J. Appl. Radiat. Isot. 18 (1967) 779.
Spencer, H., Lewin, I., Samachson, J.: Lancet 2 (1967) 156.
Carr, T.E.F., Harrison, G.E.,Humphreys, E.R., Sutton, A.: Int. J. Radiat. Biol. 14 (1968) 225.
Harrison, G.E., in: Diagnosis and treatment of deposited radionuclides, Kornberg, H.A.,
Norwood, W.D. (eds.): Excerpta Med. (1968) 333.
National Academy of Sciences: The halothane study, Washington DC: National Research
Council, 1969.
Smith, H., in: Diagnosis and treatment of deposited radionuclides, Kornberg, H.A., Norwood,
W.D. (eds.): Excerpta Med. (1968) 372.
Spencer, H., in: Diagnosis and treatment of deposited radionuclides, Kornberg, H.A.,
Norwood, W.D. (eds.), Excerpta Med. (1968) 489.
Volf, V., in: Diagnosis and treatment of deposited radionuclides, Kornberg, H.A., Norwood,
W.D. (eds.), Excerpta Med. (1968) 355.
Woodard, H.Q., in: Diagnosis and treatment of deposited radionuclides, Kornberg, H.A.,
Norwood, W. . (eds.), Excerpta Med. (1968)361.
Schofield, G.B., in: Proceedings of Symposium on Handling of Radiation Accidents, Vienna:
IAEA, 1969, p. 163.
Spencer, H., Lewin. I., Belcher, M.J., Samachson, J.: Int. J. Appl. Radiat. Isot. 20 (1969) 507.
Lambert, B.E., Sharpe, H.B.A., Dawson, K.B.: Am. Indust. Hyg. Assoc. J. 32 (1971) 682.
Vanderborght, O., Keslev, D., Van Puymbroeck, S.: Environ. Physiol. (1971) 119.
Vanderborght, O., Van Puymbroeck, S., Colard, J.: Health Phys. 21 (1971) 181.
Henry, P., in: Assessment of radioactive contamination in man, STI/PUB/290. Vienna:
IAEA, 1972, p. 641.
Humphreys, E.R., Van Puymbroeck, S., Vanderborght, O.: 2nd Int. Conf. Strontium
Metabolism, Glasgow and Strontian, USAEC Conf. 72 0818, 1972, p. 309.
Humphreys, E.R., Howells, G.R.: 2nd Int. Conf. Strontium Metabolism, Glasgow and
Strontian, USAEC Conf. 72 0818, 1972, p. 315.
International Commission on Radiological Protection: Publication 19. Oxford: Pergamon
Press, 1972.
Jolly, J., McClearen, H.A., Poda., G.A., Walke,W.P.: Health Phys. 23 (1972) 333.
Kesley, D., Van Puymbroeck, S., Vanderborght, O.: Experientia 28 (1972) 524.
Vanderborght, O.L., Colard, J., Boulenger, R.: Health Phys. 23 (1972) 240.
Schofield, G.B., Lynn, J.C.: Health Phys. 24 (1973) 317.
Schofield, G.B., Howells, H., Ward, F.A., Lynn, J.C., Dolphin, G.W.: Health Phys. 26 (1974)
541.
Spoor, N.L.,Hursh, J.B., in: Uranium, Plutonium, Transplutonium elements, Berlin: SpringerVerlag, 1973, p. 241.
Ohlenschlager, L., Schieferdecker, H., in: Diagnosis and treatment of incorporated
radionuclides, Vienna: IAEA, 1976.
Ballou, J.E., Dagle, G.E., McDonald, K.E., Buschbom, R.L.: Health Phys. 32 (1977) 479.
Spoor, N.L.: Report R-59. Chilton: National Radiological Protection Board, 1977.
Van Barneveld, A.A., Van Puymbroeck, S., Vanderborght, O.: Health Phys. 33 (1977) 533.

Landolt-Brnstein
New Series VIII/4

9-32
78S1
78V1
79S1
80K1
80M1
80N1
80P1
80S1
80T1
80V1
82S1
83B1
83M1
84W1
84S1
85M1
85S1
85S2
86L1
87B1
87S1
88G1
88T1
89B1
89C1
89D1
89D2
89L1
89N1
89S1
89S2
90D1

9 Decorporation of radionuclides
Stradling, G.N., Loveless, B.W., Ham, G.J., Smith, H.: Health Phys. 35 (1978) 229.
Volf, V.: Technical Report Series No 184, Vienna: IAEA, 1978.
Stather, J.W., James, A.C., Brightwell, J., Strong, J.C., Rodwell, P., in: Biological
implications of radionuclides released from nuclear industries, Vienna: IAEA, 1978, p. 37.
Kestens, L., Schoeters, G., Van Puymbroeck, S., Vanderborght, O.: Health Phys. 39 (1980)
805.
Metivier, H., Masse, R., Rateau, G., Lafuma, J.: Health Phys. 38 (1980) 769.
National Council on Radiation Protection and Measurements. Management of persons
accidentally contaminated with radionuclides. Report 65. Bethesda, MD: NCRP Publications,
1980.
Poda, G.A., in: The medical basis for radiation accident preparedness, Hubner, K.F., Fry,
S.A. (eds.), New York: Elsevier North Holland, 1980, p. 327.
Smith, H., James, A.C., Stradling, G.N., in: Pulmonary toxicology of respirable particles,
Springfield, VA: National Technical Information Service, 1980, p. 558.
Taylor, D.M., Volf, V.: Health Phys. 38 (1980) 147.
Voelz, G.L., in: The medical basis for radiation accident preparedness, Hubner, K.F., Fry,
S.A. (eds.), New York: Elsevier North Holland, 1980, p. 311.
Stather, J.W., Stradling, G.N., Smith, H., Payne, S., James, A.C., Strong, J.C., Ham, S.,
Sumner, S., Bulman, R.A., Hodgson, A., Towndrow, C., Ellender, M.: Health Phys. 42
(1982) 520.
Breitenstein, B.D.: Health Phys. 45 (1983) 855.
Mewhinney, J.A., Diel, J.H.: Health Phys. 45 (1983) 39.
World Health Organisation: Radiation accidents; management of overexposure. Collection
No 84-03, Geneva:WHO, 1984.
Stradling, G.N., Stather, J.W., Sumner, S.A., Moody, J.C., Strong, J.C.: Health Phys. 46
(1984) 1296.
Ma, R., Jin, Y., Zhou, Y., in: Assessment of radioactive contamination in man, Vienna:
IAEA, 1985, p. 499.
Stather, J.W., Stradling, G.N., Gray, S.A., Moody, J.C., Hodgson, A.: Hum. Toxicol. 4
(1985) 573.
Stradling, G.N., Stather, J.W., Sumner, S.A., Moody, J.C., Hodgson, A.: Health Phys. 49
(1985) 499.
Lloyd, D.C., Edwards, A.A., Prosser, J.S., Auf der Maur, A., Etzweiler, A., Weickhardt, U.,
Gssi, U., Geiger, L., Noelpp, U., Rsler, H.: Radiat. Prot. Dosim. 15 (1986) 191.
Breitenstein, B.D., Fry, S.A., Lushbaugh, C.C., in: The medical basis for radiation accident
preparedness. New York: Elsevier Science, 1987, p. 397.
Stradling, G.N., Stather, J.W., Gray, S.A., Moody, J.C., Bailey, M.R., Hodgson, A., Collier,
C.G.: Hum. Toxicol. 6 (1987) 365.
Guilmette, R.A., Muggenburg, B.A.: Int. J. Radiat. Biol. 53 (1988) 251.
Tang Minh-hua, Gong, Yi-fen, Shen Cheng-yao, Ye Chang-quing, Wu De-chang: J. Radiol.
Prot. 8 (1988) 25.
Breitenstein, B.D., Palmer, H.E.: Radiat. Prot. Dosim. 26 (1989) 317.
Carbaugh, E.H., Decker, W.A., Swint, M.J.: Radiat. Prot. Dosim. 26 (1989) 345.
Diamond, G.L.: Radiat. Prot. Dosim. 26 (1989) 26.
Durbin, P.W., White, D.L., Jeung N., Weitl, F.L., Uhlir, L.C., Jones, E.S., Bruenger, F.W.,
Raymond, K.N.: Health Phys. 56 (1989) 839.
Leggett, R.W.: Health Phys. 57 (1989) 365.
Nolibe, D., Metivier, H., Masse, R., Chretien, J.: Radiat. Prot. Dosim. 26 (1989) 337.
Stradling, G.N., Stather, J.W., Gray, S.A, Moody, J.C., Hodgson, A., Collier, C.G.: Report
M-162, Chilton: National Radiological Protection Board, 1989.
Stradling, G.N., Stather, J.W., Gray, S.A., Moody, J.C., Ellender, M., Hodgson, A., Volf, V,
Taylor, D.M., Wirth, P., Gaskin, P.W.: Int. J. Radiat. Biol. 56 (1989) 503.
Dresow, B., Neilson, P., Heinrich, H.C.: Z. Naturforsch. 45 (1990) 676.
Landolt-Brnstein
New Series VIII/4

9 Decorporation of radionuclides
90M1
90T1
91I1
91N1
91S1
92S1
92B1
92G1
93D1
93G1
93I1
93M1
93P1
93S1
93S2
94A1
94G1
94I1
94M1
94M2
94S1
94U1
94W1
95D1
95G1
95H1
95I1
95P1
95S1
95S2

9-33

Mettler, F.A., Kelsey, C.A., Ricks, R.C.: Medical management of radiation accidents. Boca
Raton: CRC Press, 1990.
Taylor, D.M., Volf, V.: Plzensky Lekarsky Sbornik (Pilsen Medical Report) Suppl. 62
(1990) 101.
International Commission on Radiological Protection: Publication 60, Oxford: Pergamon
Press, 1991.
Neilson, P., Dresow, B., Fischer, R., Heinrich, J.C.: Int. J. Appl. Instrum. 18 (1991) 821.
Stradling, G.N., Moody, J.C., Gray, S.A., Ellender, M., Hodgson, A.: Human Toxicol. 10
(1991) 15.
Stradling, G.N., Gray, S.A., Ellender, M., Moody, J.C.,Hodgson, A., Pearce, M., Wilson, I.,
Burgada, R., Bailly, T., Leroux, Y., El Manouni, D., Raymond, K.N., Durbin, P.W.: Int. J.
Radiat. Biol. 62 (1992) 487.
Bhattacharyya, M.H., Breitenstein, B.D., Metivier, H., Muggenburg, B.A., Stradling, G.N.,
Volf, V.: Radiat. Prot.Dosim. 41 (1) (1992) 1.
Gray, S.A., Stradling, G.N., Pearce, M., Moody, J.C., Ebetino, F.: Report M-339, Chilton:
National Radiological Protection Board, 1992.
Dresow, B., Neilson, P., Fischer, R., Pfau, A.A., Heinrich, H.C.: J. Toxicol. Clin. Toxicol. 31
(1993) 56.
Guilmette, R.A, Muggenburg, B.A.: Int. J. Radiat. Biol. 53 (1988) 251.
International Commission on Radiological Protection: Publication 67, Oxford: Elsevier
Science Ltd, 1993.
Moody, J.C., Davies, C.P., Stradling, G.N.: Report M-435, Chilton: National Radiological
Protection Board, 1993.
Poncy, J.L., Rateau, G., Burgada, R., Bailly, T., Leroux, Y., Raymond, K.N., Durbin, P.W.,
Masse, R.: Int. J. Radiat. Biol. 64 (1993) 431.
Stradling, G.N., Gray, S.A., Ellender, M., Pearce, M., Wilson, I., Moody, J.C., Hodgson, A.:
Human Toxicol 12 (1993) 233.
Stradling, G.N., Gray, S.A., Moody, J.C., Pearce, M., Wilson, I., Burgada, R., Leroux, Y.,
Raymond, K.N., Durbin, P.W.: Int. J. Radiat. Biol. 64 (1993) 134.
Ansoborlo, E., Henge-Napoli, M.-H., Donnadieu-Claraz, M., Roy, M., Pihet, P.: Radiat. Prot.
Dosim. 53 (1994) 163.
Gray, S.A., Stradling, G.N., Wilson, I., Moody, J.C., Burgada, R., Durbin, P.W., Raymond,
K.N.: Radiat. Prot. Dosim. 53 (1994) 319.
International Commission on Radiological Protection: Publication 66, Oxford: Elsevier
Science Ltd, 1994.
Melo, D.R.,Lipzstein, J.L., Oliveira, C.A., Bertelli, L.: Health Phys. 66 (1994) 245.
Moody, J.C., Stradling, G.N., Britcher, A.R.: Radiat. Prot. Dosim. 53 (1994) 169.
Stradling, G.N.: Radiat. Prot. Dosim. 53 (1994) 297.
Ubios, A.M., Braun, E.M., Cabrini, R.L.: Health Phys. 66 (1994) 540.
Wood, R., Britcher, A.R, McGinn, J., in: Proceedings of Regional Conference of IRPA,
Portsmouth, UK, Ashford: Nuclear Technology Publishing, 1994, p. 165.
Dean, M.R.: An evaluation of the use of broncopulmonary lavage in the treatment of
plutonium oxide, Report NRPC 54, Greenwich: Royal Naval College, 1995.
Gray, S.A., Pearce, M.J., Stradling, G.N., Wilson, I., Hodgson, A., Isaacs, K.R.: Human
Toxicol. 14 (1995) 902.
Henge-Napoli, M.-H., Archimbaud, M., Ansoborlo, E., Metivier, H., Gourmelon, P.: Int. J.
Radiat. Biol. 68 (1995) 389.
International Commission on Radiological Protection: Publication 69, Oxford: Elsevier
Science, 1995.
Paquet, F., Poncy, J.L., Metivier, H., Grillon, G., Fritsch, P., Burgada, R., Bailly, T.,
Raymond, K.N., Durbin, P.W.: Int. J. Radiat. Biol. 68 (1995) 663.
Stradling, G.N., Moody, J.C.: Radioanalyt. Nucl. Chem. Articles 197 (1995) 309.
Stradling, G.N., Gray, S.A., Pearce, M., Wilson, I., Moody, J.C., Burgada, R., Durbin, P.W.,
Raymond, K.N.: Human Toxicol. 14 (1995) 165.

Landolt-Brnstein
New Series VIII/4

9-34
96H1
96I1
96O1
96V1
97S1
98A1
98B1
98D1
98G1
98H1
98I1
98M1
98S1
98S2
99H1
99W2
00R1
00G1
00H1
00M1
00P1
00S1
00S2
00S3
00W1

9 Decorporation of radionuclides
An American National Standard - Bioassay Programs for Uranium. Report HPSN.22-1995.
McLean VA: Health Phys. Soc. 1996.
International Commission on Radiological Protection: Publication 71, Oxford: Elsevier
Science Ltd, 1996.
Official Journal of the European Communities: L159, Vol. 39, 29 June 1996, Luxembourg:
Office for Official Publications of the European Community, 1996.
Volf, V., Burgada, R., Raymond, K.N., Durbin, P.W.: Int. J. Radiat. Biol. 70 (1996) 109.
Stradling, G.N., Hodgson, A., Fell, T.P., Rance, E.R.: Report M-801, Chilton: National
Radiological Protection Board, 1997.
Ansoborlo, E., Hodgson, A., Stradling, G.N., Hodgson, S., Metivier, H., Henge-Napoli, M.H., Jarvis, N.S., Birchall, A.: Radiat. Prot. Dosim. 79 (1998) 23.
Bailly, T., Burgada, R.: Comptes Rend. Acad. Sci. Paris t.1, Ser. IIc (1998) 241.
Durbin, P.W., Kullgren, B., Xu, J., Raymond, K.N.: Radiat. Prot. Dosim. 79 (1998) 433.
Gourmelon, P.: Unpublished data.
Henge-Napoli, M.-H., Ansoborlo, E., Houpert, P., Mirto, H., Paquet, F., Burgada, R.,
Hodgson, S.,Stradling, G.N.: Radiat. Prot. Dosim. 79 (1998) 449.
International Atomic Energy Agency: Dosimetric and medical aspects of the radiological
accident in Goiania in 1987. IAEA-TECDOC-1009, Vienna: IAEA, 1998.
Madshus, K., Stromme, A., Bohne, F., Nigrovic, V.: Int. J. Radiat. Biol. 10 (1966) 519
Stradling, N.,Hodgson, S.A., Pearce, M.: J. Radiat. Prot. Dosim. 79 (1998) 445.
Stradling, G.N.: J. Alloys Compounds. 271-273 (1998) 72.
Harrison, J.R., Paile, W., Baverstock, K.: Radiation and cancer , Singapore: World Scientific,
EUR 18552, 1999, p. 455.
Guidelines for iodine prophylaxis following nuclear accidents, update 1999, Geneva, World
Health Organization, 1999.
Rote Liste: Bundesverband der Pharmazeutischen Industrie eV, Frankfurt, 2000.
Geoffroy, B.,Verger, P., Le Guen, B.: Radioprotection 35 (2) (2000) 151.
Henge-Napoli, M.-H., Stradling, G.N., Taylor, D.M. (eds): Radiat. Prot. Dosim. 87 (2000) 1.
Martinez, A.B., Cabrini, R.L., Ubios, A.M.: Health Phys. 78 (2000) 668.
Phipps, A.: NRPB, personal communication, 2000.
Stradling,G.N., Henge-Napoli, M.-H.,Paquet, F., Poncy, J.-L.,Fritsch, P.,Taylor, D.M.:
Radiat. Prot. Dosim. 87 (2000) 19.
Stradling, G.N., Henge-Napoli, M.-H., Paquet, F., Poncy, J.-L.,Fritsch, P.,Taylor, D.M.
Radiat. Prot. Dosim. 87 (2000) 29.
Stradling, G.N., Taylor, D.M., Henge-Napoli, M.-H., Wood, R., Silk, T.J.: Radiat. Prot.
Dosim. 87 (2000) 41.
Wood, R., Sharp, C., Gourmelon, P., Le Guen, B., Stradling, G.N., Taylor, D.M, HengeNapoli, M.-H.: Radiat. Prot. Dosim. 87 (2000) 51.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-25]

10 Measuring techniques

10-1

10 Measuring techniques

Since human beings do not possess any sense for the detection of ionizing radiation, they must entirely
rely on special instruments in order to prevent or control any harmful radiation exposure or intake of
radioactivity from the outset. Therefore, reliable instruments and methods for radiation detection and
measurement form the precondition for the safe handling of radiation and radioactivity in medicine,
scientific research, industry, and nuclear energy production. As summarized in Table 10.1, the primary
tasks of radiation protection measurements can roughly be divided into dose and activity measurements
employed to prevent and control hazard to man from ionizing radiation (external exposure) or the incorporation of radioactivity (internal exposure), respectively. The purpose of this Chapter is to give a short
overview of radiation detectors frequently utilized for these tasks (10.1) and of their application in practice to monitor and quantify external (10.2) and internal exposures (10.3).

10.1 Detectors for radiation protection


10.1.1 Overview and general characteristics of radiation detectors
The function of a detector designed of measuring ionizing radiation is to generate a measurable response
that is related to the energy deposited in the detector material or the number of particles entering into it
such as the charge produced in a gas, the intensity of visible light emitted by some solid or liquid matter,
the degree of blackening of a photographic film, or the number of chromosome aberrations in a biological
sample. The most common radiation effects used for radiation protection measurements are summarized
in Table 10.2. In this Chapter (10.1) the emphasis is on how these detectors and the instruments based
on them respond to radiation and how this response can be interpreted to determine the desired quantity
such as dose, exposure, or activity. Clearly, radiation protection measurements are performed by means of
instruments common to other fields of radiation detection and measurement (e.g. nuclear and particle
physics, radiation therapy, diagnostic radiology, and nuclear medicine). Therefore, much has been published about the design and operation of these devices. An enormous amount of information is provided,
among others, in the excellent textbooks by Attix [86Att], Kiefer and Maushart [72Kie], Knoll [00Kno],
Leo [94Leo], Lutz [99Lut], Shani [00Sha], Tsoulfanides [95Tso], and in the comprehensive three-volume
treatise edited by Attix, Roesch, and Tochilin [68Att]. These texts form the basis of the following brief review and the reader is referred to them for supplementary information and an exhaustive bibliography.

Landolt-Brnstein
New Series VIII/4

10-2

10 Measuring techniques

[Ref. p. 10-25

Table 10.1. Major objectives and categorization of radiation protection measurements.


Objective of
External exposure
Internal exposure
Measurement
Prevention of
exposure

Control of
Exposure

Area monitoring
Assessment of protective
measures

Personal monitoring

Determination and identification


of radioactivity in air, water, and
food that may be incorporated
Environmental monitoring
Detection of surface or skin
contaminations
Determination and identification
of incorporated radioactivity by in
vivo and excretition measurements

Table 10.2. Radiation effects frequently used for radiation protection measurements (adapted from
[96Hoh]).
Radiation effect Type of instrument
Detector material
Determina- Determina- Identification of dose tion of
tion of
or dose rate activity or radioparticle
nuclides
counting
Ionization chamber
Gas
++
+
+
Electrical
Proportional counter Gas
+
++
++
Geiger-Mller counter Gas
+
++
Semiconductor
Crystalline
++
++
++
detector
semiconductor
Optical
Scintillation counter Crystal, plastic, or liquid ++
++
++
Thermoluminescence Crystal
++
dosimeter
Radiophotolumines- Glas
+
cence dosimeter
Thermal
Superheated drop
Liquid drops
+
detector
Chemical
Film
Photographic emulsion ++
Etched-track detector Plastic foil
+
Biological
Scoring of chromoChromosomes
+
somal aberrations
++ : frequently employed, recommended procedure
+ : in specific cases usefully applicable
Before the different radiation detectors mentioned in Table 10.2 are described in more detail in the
sub-sequent Sections, two fundamental aspects which are relevant for all of them are outlined [68Att,
86Att]:
Absolute measurement and calibration. A device is regarded as absolute if it can be constructed and
subsequently used to measure radiation without the necessity of calibrating its response in a known radiation field. Such instruments are primarily installed and operated in national standards laboratories. For
radiation protection measurements it is sufficient to use devices that are properly calibrated by accredited
laboratories that guarantee traceability to an official standards laboratory. Calibration can be accomplished, for example, by exposing a device (in some cases in or in front of an appropriate phantom) in a
reference radiation field, and then determining a calibration factor N by which the detector reading R has
Landolt-Brnstein
New Series VIII/4

Ref. p. 10-25]

10 Measuring techniques

10-3

to be multiplied to achieve the true value of the measurand. If the quantity to be measured is not linearly
related with detector response, a calibration curve has to be established by varying the considered parameter of the radiation field over the relevant range. It is important to note, that detector response generally
depends on the type and energy of the radiation (photons or particles) to be detected. Due to this reason,
calibration of a radiation measuring instrument is restricted to a specified range of both the quantity to be
determined and the energy of the radiation considered.
Repeatability, accuracy, and uncertainty. The repeatability of an instrument is its ability to yield the
same result for repeated measurements in a constant radiation field. Consequently, it can be stated in
terms of the standard deviation estimated from the readings of repeated measurements. On the other hand,
the accuracy of an instrument expresses the closeness of the reading to the true value of the quantity being
measured and, thus, mainly depends on the correctness of its calibration. In other words: repeatability
characterizes random errors due to instabilities of the instrument and the stochastic character of ionizing
radiation, whereas accuracy quantifies systematic errors of the measurement process. In general, the result
of a measurement is only an approximation of the true value of the measurand, and thus is complete only
when accompanied by a quantitative statement of its uncertainty, i.e. an interval within which the true
value lies with given probability [95ISO]. Possible sources of error in radiation dosimetry and procedures
for estimating the resulting magnitude of the uncertainties in the measurement results can be found in
[02ISO].

10.1.2 Gas-filled ionization detectors


10.1.2.1 Ionization and gas amplification
Ionization detectors were the first electrical devices developed for radiation detection. They are still
widely used for radiation protection measurements. Radiation detection is based on the collection of electrons and ions produced by ionizing particles in the detector material preferential a gas because of the
high mobility of electrons and ions in this medium. An atom can be ionized in a variety of ways, but
collisions of charged particles with the atomic electrons via Coulomb interaction are the most important
mechnisms. For most gases of practical interest, the average energy W spent by a charged particle to
produce an electron-ion pair is between 20 and 40 eV [79ICR]. In air, for example, the value is about 36
and 34 eV for and particles, respectively. Consequently, a charged particle will produce a large
number of electron-ion pairs if it deposits its energy completely in the gas.
As shown in Fig. 10.1, a typical detector configuration consists of an anode wire inside of a cylindrical cathode. When an electric potential difference (voltage) is applied between the electrodes, electrons
and ions are attracted to the electrodes and generate an electric output signal, which is passed through a
series of electronic circuits for amplification, processing, and storage. There are two different ways to
process the signal. In the pulse mode, the electrical signal from each event is processed individually,
whereas in the current mode, the signals from individual interactions are integrated, yielding a net current
signal. Depending on the voltage between the electrodes, there are three basic types of gas-filled
ionization detectors taking advantage of different physical effects: the ionization chamber, the proportional chamber, and the Geiger-Mller counter. To understand the differences between these devices,
consider under the assumption of a constant flux of radiation the behavior of a gas-filled ionization
detector as the electrical voltage is increased. As illustrated in Fig. 10.2 for two different types of particles, six different regions of operation can be distinguished [00Kno, 94Leo]:
At very low voltages, the electric field is insufficient to avoid recombination of the electron-ion pairs.
However, recombination is lessened as the voltage is raised, giving rise to a saturation curve (region I).
Usually, the saturation curve reaches a more or less distinct plateau, where the number of ions collected is
nearly independent of the applied voltage. A detector operating in this region (II) is denoted as ionization
chamber.

Landolt-Brnstein
New Series VIII/4

10-4

10 Measuring techniques

[Ref. p. 10-25

As the voltage across the device is raised further, the number of ions collected starts to increase again,
because the electric field is strong enough to accelerate electrons from the primary ionization to kinetic
energies that are sufficiently high to ionize further gas molecules by collisions (secondary ionization). In
the case of a cylindrical detector arrangement, the electrical field increases steeply near the anode wire
and thus the amplification process results in an electron avalanche confined to a very small length of the
anode wire. Due to this reason, electron avalanches initiated by primary ionizations at different points in
the gas do not interfere and thus the output of the device is for a given voltage directly proportional to
the number of primary ionizations or, equivalently, to the energy dissipated inside the detector volume. A
detector operating in this region (III) is known as a proportional counter. Increasing the applied voltage
still more results in a nonlinear amplification process (region IV) since the gain is so large that spacecharge effects become relevant distorting the electrical field within the detector.
If the voltage between the electrodes exceeds the region of limited proportionality, the number of ions
collected becomes independent of the type and energy of the incident particles. The reason is that instead
of various independent avalanches confined to small Sections along the wire (as in regions III and IV), a
chain reaction of many avalanches is triggered, which spread out along the entire wire. This transitory
electrical breakdown is initiated by excited gas atoms undergoing radiative de-excitation. Ultraviolet
photons emitted by these processes propagate to other parts of the gas volume and initiate secondary
avalanches. Detectors working in this region (V) are called Geiger-Mller counters. As the voltage is
further increased, a continuous breakdown occurs (region VI).
E ( r ) ~1/r
to electrometer
or amplifier

Fig. 10.1 Typical arrangement of a cylindrical ionization detector operated in the current mode. (r, radial distance from the
anode wire; E, transaxial electric field strength).

Number of ions collected per time


(logarithmic scale)

particle
particle

II

III

IV

VI

Fig. 10.2 Schematic plot showing the relation between the


voltage applied to a gas-filled ionization detector and the
charge collected for two different types of particles
depositing different amounts of energy within the ioncollecting gas volume. Six regions of operation can be
distinguished: I, recombination region; II, ionization
region; III, proportional region; IV, limited proportional
region; V, Geiger-Mller region; VI, discharge region.

Applied voltage

10.1.2.2 Ionization chambers


Ionization chambers are frequently used for radiation protection measurements and are available in a
large variety of types. They are normally used in the current and not in the pulse mode. The electrical
current caused by a device operating in the ionization region, however, is much too small to be measured
using conventional galvanometer techniques. Therefore, a sensitive electrometer with a sufficiently high
impedance indirectly measures the electrical current in the circuit by detecting the voltage drop across a
high-load series resistor (cf. Fig. 10.1 and [00Kno]). Depending on whether the charge or the current is
measured, the chamber will register the total ionization that has occurred in a given time (exposure, dose)
or the rate of ionization at any instant (exposure rate, dose rate).
Landolt-Brnstein
New Series VIII/4

Ref. p. 10-25]

10 Measuring techniques

10-5

To discuss the basic concepts, we first consider free-air ionization chambers, which are in operation
primarily in national standards laboratories. They form the standard against which simpler instruments are
compared to yield reliable calibration factors. A typical configuration of a parallel-plate ionization
chamber is shown in Fig. 10.3. An important feature of this device is that the sensitive volume, i.e. the
volume common to both the collecting volume defined by the electrical field between the plates and the
collimated beam (cf. Fig. 10.3), is defined electronically and not by the walls of the chamber. Free-air
chambers are specifically well-suited for the measurement of radiation exposure since this quantity
according to the operational definition given in Section 4.4.2.3 is directly related to the ionization
produced by collision interactions in air by charged particles resulting from interactions of photons (or
neutrons). However, exposure measurements require the detection of all the ionization produced in the
selecting volume. To this end, the lateral distance between each of the electrodes and the border of the
sensitive volume must exceed the range of the secondary electrons which originate in the sensitive
volume. This guarantees that electrons, the path of which remains within the collecting volume (like e1 in
Fig. 10.3), can produce all their ionization in this region where it will be collected and measured. But
most electrons released in the sensitive volume will leave the collecting volume (like e2) and thus part of
the ionization they produce will not reach the collector. This loss, however, is of no relevance if as many
electrons from photon interactions elsewhere in the beam enter the sensitive volume with the same energy
(like e3) as leave it. A detailed discussion of this condition, which is known as charged particle equilibrium (or in the considered case of a photon beam as electronic equilibrium), can be found in [86Att].
Charged particle equilibrium in the sensitive volume is attained when the photon flux remains constant
across the chamber and when the distance from the diaphragm of the chamber to the border of the sensitive volume is greater than the maximum electron range. Since the maximum range of electrons in air increases rapidly with the energy of incident X- or -rays, the size of a free-air ionization chamber that can
be realized in practice limits the energy of photons to be measured to about 200 keV.
to electrometer
Collector

Guard
electrodes
Diaphragm
Source

Wires

e1
e2

e3

Sensitive
volume

Collecting
volume

X or
rays

Fig. 10.3 Typical configuration of


a parallel-plate free-air ionization
chamber. The grounded guard
electrodes and the electrically
biased wires are used to produce a
uniform electrical field within the
collecting volume. The ionization
measured is that produced by
electrons in the collecting volume.

To avoid the use of large and cumbersome free-air ionization chambers, practical cavity ionization
chambers have been developed which basically consist of a solid wall surrounding a gas-filled cavity. In
this cavity an electric field is established to collect the ions produced by radiation entering the chamber
(cf. Fig. 10.1). The criteria that determine the dimension and the material of a chamber depend on both
the desired dosimetric quantity (exposure or absorbed dose) and the type and energy of the radiation to be
measured. Cavity ionization chambers can be designed and operated either as equilibrium or Bragg-Gray
chambers in which the local photon (or neutron) field or the local secondary charged-particle field is sufficiently well characterized, respectively [86Att].
Dosimetric measurements in photon and neutron fields can be performed with cavity chambers operated under charged particle equilibrium conditions. The physical basis is described by the Fano theorem
which states that in an infinite medium of given atomic composition exposed to a uniform field of indirectly ionizing radiation, the field of secondary radiation is also uniform and independent of the density of
the medium as well as of density variations from point to point [86Att]. That means that the ionization
Landolt-Brnstein
New Series VIII/4

10-6

10 Measuring techniques

[Ref. p. 10-25

collected in the gas is not influenced by the density of the wall, provided that the materials of the wall and
the gas are matched with regard to their atomic composition i.e. the effective atomic number Z and
that the thickness of the wall exceeds the range of the charged secondary particles released in the wall.
The dimensions of the gas-filled cavity can be adapted to the flux of the radiation field to be measured in
order to generate a sufficiently high ionization. For exposure measurements, the wall should be made of
air-equivalent material with an effective atomic number of air, such as some plastics or graphite
with silicon additives. Although there is no material that is exactly air-equivalent, the deviations are of
minor importance for the purposes of radiation protection measurements. The use of cavity chambers
operated under charged particle equilibrium conditions is limited to photons with an energy of less than
about 3 MeV. At higher energies it is not possible to build a chamber that meets the two central conditions, namely a sufficiently thick wall to establish electronic equilibrium and negligible attenuation of the
photon beam crossing the chamber. Chambers can also be made tissue-equivalent, which means that the
atomic composition of both the wall and the gas must be similar to that of tissue. Since the absorbed dose
in air or tissue (at a depth corresponding to the wall thickness) is directly proportional to the ionization
per unit volume of gas measured under charged particle equilibrium conditions with an air- or tissueequivalent chamber, respectively, these devices can be calibrated to directly read the desired dosimetric
quantity.
Dosimetric measurements can also be performed with cavity ionization chambers taking advantage of
the Bragg-Gray principle, which states that the absorbed Dose Dm in a given homogeneous medium can
be calculated from the ionization produced in a small gas-filled cavity suspended into this material according to the equation Dm = WS m g J , where W is the average energy dissipated in the gas per electronion pair formed, S m g the ratio of the average mass stopping powers of the medium and the gas for the
charged particles considered, and J the ionization per unit volume of gas. This principle holds under two
conditions: Firstly, the cavity must be small compared to the range of the charged particles striking so that
the flux and energy spectrum of the charged particles is not disturbed. Secondly the energy lost by the
charged particles in crossing the gas-filled cavity is equal to the energy deposited in the gas volume
[86Att]. In practice, the gas-filled cavity is part of a cavity chamber with a wall that separates the cavity
from the surrounding material. Therefore, the chamber wall should be either extremely thin to leave the
charged particle field unchanged or matched to the atomic composition of the surrounding medium.
Bragg-Gray cavity chambers can be used for dosimetry of charged particles, entering from outside the
chamber, and of high-energy photons (>3 MeV) liberating electrons in the chamber wall.
In practice, neither the idealized conditions of charged particle equilibrium nor that of the Bragg-Gray
principle can be fully realized and thus a variety of corrections must be applied in order to make absolute
dosimetric measurements possible. For radiation protection measurements, however, this complex task
can be avoided when chambers are used that are calibrated under conditions similar to those in which the
instrument will be applied and in terms of the desired dosimetric quantity (e.g., exposure, absorbed dose,
ambient dose equivalent, or the corresponding dose rates). As an example, Fig. 10.4 shows a portable
survey instrument with an air-equivalent chamber that can be used for dose and dose rate measurements
over a wide range of photon energies.

Fig. 10.4 Dose and dose rate survey meter with an air- equivalent
ionization chamber (volume 600 cm3) for the measurement of X- and
-rays in the energy range between 6 keV and 3 MeV. Using an
additional plastic shielding, even photons up to an energy of 7.5 MeV
can be measured. The probe is detachable for remote measurements.
(Courtesy Step Sensortechnik).
Landolt-Brnstein
New Series VIII/4

Ref. p. 10-25]

10 Measuring techniques

- - - -

Charging
electrode

+ + + +
+ + + +

- - - -

10-7

C
Insulator

Fig. 10.5. Schematic representation of a condenser-type


ionization chamber.

Other types of portable ion chambers are based on the charge integration principle illustrated in
Fig. 10.5. In this case, the chamber electrodes are connected in parallel with a capacitor, which is initially
charged up to establish an electric field in the chamber. When the chamber is exposed to radiation, the
ionization caused in the gas-filled chamber is collected by the electrodes and the charge stored in the capacitor is reduced. The resulting drop in chamber voltage can be used as a measure of the total integrated
ionization charge. In the case of a self-reading pocket dosimeter employed for personal radiation protection measurements, a simple rugged quartz-fiber is mounted inside the ionization volume which is
deflected when the device is charged. The position of the fiber, which varies as the charge on the capacitor is reduced due to ionizing radiation, is observed through a small built-in microscope that has a scale in
the eyepiece (Fig. 10.6). The position against the scale can be calibrated in terms of the total radiation
recorded by the pocket dosimeter. Over a longer period of time, however, leakage currents across the insulator surface can not be avoided and this limits the accuracy of the dosimeter.
Progress in integrated circuit technology resulted in the development of direct ion storage (DIS) dosimeters, which combine a gas-filled ionization chamber with a non-volatile electronic charge storage element [96Kah, 00Kno, 99Wer]. A schematic representation of a DIS dosimeter is shown in Fig. 10.7. The
charge is initially placed on the floating gate of a standard EEPROM (electrically erasable and programmable read-only memory) cell by injecting electrons by a tunneling process through the silicon
oxide. Electrons are trapped at this gate for many years since at normal operating temperatures they have
no conductive discharge path when the silicon dioxide formation is made of high-purity material (cf.
Section 10.1.4). When the chamber is exposed to ionizing radiation, the ions produced in the fill gas are
collected by the charged floating gate, which results in a reduction of the charge stored. Assessment of
this quantity can be performed without changing the charge distribution by measuring the channel conductivity of the field effect transistor (FET), which forms the basis of the EEPROM, by means of an
electronic readout unit. The passive electronic dosimeter thus makes it possible to instantly and nondestructively readout the accumulated dose whenever required. The first commercial DIS personal
dosimeter consist of a series of separate dosimeter elements housed inside a small hermetically sealed
container. Three elements are used for the measurement of the personal dose equivalent Hp(10) in the
range from 1 Sv to 40 Sv and two elements for the determination of Hp(0.07) in the range between
10 Sv and 40 Sv [99Wer] (the definition of Hp(d) is given in Section 4.5.3.4). The dose-rate linearity is
flat up to 40 Sv/h thus guaranteeing accurate dose assessment in accident situations.
Field lens

Eye lens

Scale

Fiber

Insulator

Ionization
chamber

Electrode

Bellows

Charging
pin

Fig. 10.6. Pen-size direct-reading ion chamber dosimeter for personal radiation protection measurements in a - or
X-ray environment. (a) Simplified schematic representation. (b) Pocket dosimeters for different dose and energy
ranges. The nominal voltage required to set the dosimeter to zero is adjusted with the charging unit (potentiometer).
A test source is used to check the correct function and calibration of the personal dosimeters. (Courtesy Thermo
Eberline ESM).
Landolt-Brnstein
New Series VIII/4

10-8

10 Measuring techniques

Silicon
oxide

Source

Fill gas

Channel

Floating gate

Drain

Silicon

[Ref. p. 10-25

Tunneling
process

Fig. 10.7. Schematic representation of a DIS dosimeter


consisting of combination of a gas-filled ionization
chamber and an EEPROM.

10.1.2.3 Proportional counters


A serious limitation of ionization chambers is that they are not sensitive enough to detect individual ionizing particles and thus can not be used for particle counting. This limitation can be overcome by operating an ionization detector in the proportional region to take advantage of the gas amplification process
described in Section 10.1.2.1. However, proportional counters are not simply ionization chambers operated at high voltages (predominantly) in the pulse mode, but are specially constructed devices designed to
optimize the gas amplification effect [00Kno]. The most important difference is that proportional counters
always contain a thin anode wire to create a high electric field. Important geometrical factors are among
others the uniformity, smoothness, and centricity of the thin anode wire with a diameter of between 5
m and 50 m. Specific demands on the gases are: low working voltage, high gas amplification, good
proportionality, and high rate capability. These conditions are met by using mixtures of a noble gas and a
polyatomic organic gas, such as 90 % argon and 10 % methane (P10 gas) or 96 % helium and 4 %
isobutane. The organic additives, denoted as quenchers, improve the stability and performance of the
counter by absorbing secondary ultraviolet photons that are emitted from excited gas atoms in a mode
that does not lead to further ionization and thus avoids a transitory electrical breakdown. In practice, gain
factors between 102 and 106 can be achieved.
For particle detection and counting, proportional counters are operated in the pulse mode. Fig. 10.8
shows the equivalent circuit of a proportional counter, which replaces the circuit plotted in Fig. 10.1 for a
current-type chamber. As mentioned above, the electrical field increases steeply near the central wire of a
ionization detector and thus the amplification process initiated by an ionizing event in the sensitive volume results in an electron avalanche in a region extending only a fraction of a millimeter from the anode
surface. However, at the moment when the electrons are collected at the anode wire (within about 1 s),
the positive ions are still so close to the center wire that there is almost no change of the electric voltage.
The output signal a voltage pulse V(t) is thus predominantly determined by the slower drift of the
positive ions outward towards the cathode. Most of this process develops while the ions are still relative
close to the wire and thus a sharply defined fast-rising electrical pulse can be observed. The subsequent
decrease of the voltage pulse depends on the relative time constant of the external load circuit, which is
given by the product of the resistance R and the equivalent capacitance C of both the detector and the
measuring circuit (usually a preamplifier). When the capacitance of the circuit is fixed, the height of the
voltage pulse is directly proportional to the charge generated within the detector and thus to the amount of
energy the incident particle deposited in the gas [00Kno].
Based on this feature, a discrimination between particles depositing different amounts of energy in the
gas volume such as - and -particles can be achieved. This can be realized, for example, by means of
two separate read-out channels with different discriminator levels (cf. Fig. 10.8) in order to simultaneously detect either -particles (high level) or both - and -particles (low level). Alternatively, the
proportional counter can be connected to a multichannel analyzer, which records and stores pulses according to their height and thus allows the direct discrimination between different particles. If the channel
number is related to the energy loss of the incident particles in the cavity by means of a suitable calibration procedure, a proportional counter can also be used for particle identification (particle spectroscopy).

Landolt-Brnstein
New Series VIII/4

10 Measuring techniques

V (t)

Voltage V ( t )

Counter

Ref. p. 10-25]

10-9

VD

Time t

Fig. 10.8. Equivalent circuit of a counter operated in the pulse mode (cf. Fig. 10.1). VD is the
discriminator level. Only pulses with a pulse height exceeding this level are counted.

In practice, it is not possible to record all types and energies of radiation with sufficient efficiency by
one detector only and thus different detector designs are used for -, -, and -radiation. For the detection
of - and low-energy -particles, it is of particular importance to use counters with thin window foils
(such as metalized plastic) or windowless entrance cathodes made of a metal wire grid to reduce absorption or energy-loss of the particles transversing the entrance material as far as achievable. Since such
systems can be sealed only heavily, stationary counters are normally operated as gas flow units to avoid
gradual contaminations and loss of fill gas. To monitor for contaminations on floors and surfaces of
objects in laboratories or on hands, shoes, and clothing of personnel working with radioactive material,
large-area proportional counters are used such as those shown in Fig. 10.9. They consist of a cathode
container filled with gas, which encloses either a meander-shaped anode wire or multiple anode wires.
Proportional counters (PC) made of a tissue equivalent (TE) plastic wall (most often A-150 plastic)
and filled with TE gas mixtures (propane or methane gases mixed with carbon dioxide and nitrogen) are
standard instruments in microdosimetry [83ICR]. An important feature of TEPCs, which are based on the
Bragg-Gray cavity theory described in Section 10.1.2.2, is that the pressure of the filling gas can be
adjusted so that a charged particle crossing the cavity deposits an identical amount of energy as a charged
particle crossing a real tissue volume of microscopic dimensions [95Wak]. Multichannel pulse-height
measurements with a low-pressure TEPC in radiation fields with an intensity low enough to allow the
detection of single events thus give the distribution of the energy deposited by individual primary charged
particles in a microscopically small tissue volume. Since the height of the recorded pulses strongly
depends on the ionization density along the tracks of the charged particles which varies considerably
between different types of charged particles such as electrons, protons and heavier ions a low pressure
TEPC not only acts as recorder of deposited energy, but also as a spectrometer able to distinguish charged
particles with a different linear energy transfer (LET) and thus provides an estimate on radiation quality
[95Wak, 02Wak].

a
Landolt-Brnstein
New Series VIII/4

Fig. 10.9. Large-area proportional


counters for contamination detection. a: Sealed handheld monitor
for detection of surface contaminations with -, - and -isotopes.
(Courtesy Thermo Eberline ESM).
b: Continuous gas-flow contamination monitor for detection of
- and -contaminations of hands,
shoes, and clothing. (Courtesy
Berthold Technologies).

10-10

10 Measuring techniques

[Ref. p. 10-25

Low pressure TEPCs have been an important laboratory tool in experimental microdosimetry for
many years. The recent availability of microelectronics and progress in digital electronics enabled the
development of portable TEPCs for area monitoring and thus the application of this microdosimetric
method in practical radiation protection [89Men, 95Sch, 00Sha]. Particular advantages of these devices
are the possibility (1) to separate dose fractions due to photons and neutrons in mixed n- fields, which
release weakly respectively densely ionizing secondaries in the TE wall of the counter, and (2) to directly
measure the operational quantity ambient dose equivalent [89Die].
10.1.2.4 Geiger-Mller counters
Although the design and operation of Geiger-Mller (GM) counters are in many respects similar to those
of proportional counters, there are three important differences: Firstly, GM counters are operated at substantially higher tube voltages (cf. Fig. 10.2) so that a particle entering the gas volume triggers an electron
avalanche extending along the entire length of the anode wire. As a consequence, the size of the output
voltage pulses is more or less independent on the number of original ion pairs that initiate the gas amplification process and thus on the type and energy of particle entering the sensitive gas volume. The voltage
between the electrodes that is required to sustain an avalanche ionization can be determined by exposing
the counter to a constant source of radiation and observing the counting rate as a function of the applied
voltage. Secondly, the UV absorbing quench gas used in proportional counters is omitted in GM tubes
since ultraviolet photons emitted from excited gas atoms are essential to the process of propagating the
discharge throughout the tube. Instead, other quenchers such as gaseous halogens (Cl or Br) or organic
substances (ethyl alcohol or ethyl formate) are added with a typical concentration of 5-10 % to the
primary fill gas to prevent repeated or continuous gas discharge through the mechanism of charge transfer
collisions. A detailed description of the underlying complex mechanism can be found in [86Att, 00Kno].
Thirdly, immediately after a discharge, a dense cloud of positive ions exists near the central wire and reduces the electric field in the counter to a great extent. This space charge not only terminates the discharge of the tube but also prevents that a further avalanche can be generated before the positive ions
have moved at least part of the distance towards the cathode. The time between the detection of the
initial pulse and the time at which a succeeding pulse can be counted because its amplitude exceeds the
discriminator level is denoted as resolving time. Typical values are between 100 to 300 s. In contrast,
the resolving time of a proportional counter is less than a few microseconds. During the resolving time,
the GM counter is dead and any particles entering the tube during that time are lost [94ICR1].
In practice, gain factors between 106 and 1010 can be realized with a GM counter. The resulting voltage pulses have a height between 1 and 10 V and can easily be detected with simple electronic circuits
often completely without external amplification. GM tubes are thus simple, rugged, and relatively inexpensive particle-counting instruments. As mentioned above, however, they suffer from extremely long
resolving times and are thus seldom used when accurate measurements are required at count rates greater
than a few hundred counts per second. In many cases, GM counters are provided with removable covers
on the entrance window in order to differentiate between penetrating (- and high-energy -particles) and
low-penetrating (- and low-energy -particles) radiation by measuring the difference between the count
rates with and without the cover in place. As in the case of proportional counters, the entrance window
must be sufficiently thin to permit passage of -particles. For the detection of -particles, on the other
hand, the thickness of the entrance window or of the cover should approximate the maximum range of the
secondary electrons produced in the window or cover to increase detection sensitivity.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-25]

10 Measuring techniques

10-11

10.1.3 Scintillation detectors


The major limitation of gas-filled counters, namely the low detection efficiency for X- or -rays, can be
overcome by the use of solid or liquid detector materials, which have a much higher density than gases.
Scintillation materials are frequently utilized for photon or neutron detection. In these materials a small
fraction of the energy deposited by charged particles will be emitted as visible or ultraviolet light on a
time scale of nanoseconds to milliseconds, whereby the intensity of the light flash is proportional to the
energy deposited in the scintillator. As shown in Fig. 10.10, a scintillation detector consists basically of
scintillator material that is optically coupled to a photomultiplier tube to convert the light photons
released in the scintillator into an electrical pulse which can then be amplified and analyzed electronically.
Scintillator

particle
Analyzer

Semitransparent
photocathode

Light shield, reflector


Photomultiplier

gas envelope

HV

Shield
e

Amplifier

Pre
amplifier

Light
photon

Dynodes

Anode

Fig. 10.10. Schematic representation of (a) a pulse-mode scintillation detector and (b) a linear focussed
photomultiplier tube showing the cascade effect due to amplification of electrons from the photocathode by increasing secondary emission when the electrons strike the dynodes.

Scintillators fall into two major categories: inorganic and organic materials, the choice of which depends strongly on the type of measurement to be performed. Physical properties of a few representative
scintillator materials are given in Table 10.3; a more comprehensive list can be found in [00Kno, 94Leo].
Inorganic scintillators are crystals of alkali halides (such as NaI, CsI) or oxides (such as Bi4Ge3O12,
BGO) grown at high temperatures. In these materials, scintillation is a property of the electronic band
structure of the crystals: When an ionizing particle enters the scintillator, it can raise electrons from the
valence into the conduction band. The electrons and holes formed by this excitation process recombine
and emit a photon. In the pure scintillator material, however, de-excitation is an inefficient process due to
self-absorption. Therefore, small amounts of an activator (e.g., thallium in the case of NaI) are added.
These impurities create energy states within the forbidden band gap of the scintillator over which electrons can alternatively de-excite from the conduction band into the valence band. A more detailed description of the scintillation process can be found in [00Kno, 95Tso]. Since energy spacing between activator energy states is less than that between the conduction and valence bands of the pure solid, the
emitted photons do not have enough energy to raise other electrons from the valence band to the conduction band and thus cannot be effectively reabsorbed by the scintillator. Moreover, the change in energy of
the emitted photons results in a shift of the wavelength of maximum emission from the ultraviolet into the
visible region, where the sensitivity of most photomultiplier tubes is maximal. Inorganic scintillators tend
to contain elements with a high atomic number and have a relatively high density (cf. Table 10.3). Consequently, the photoelectric effect is the main interaction mechanism for X- or -rays in the energy range
between 10 keV to 1 MeV, making inorganic scintillators favorable for particle identification by means of
spectroscopic measurements (see below). They also have a high light output, but are hampered by a
relatively slow response.
Organic scintillators, on the other hand, are aromatic hydrocarbon compounds which contain benzenoid rings. They are broadly classed into three types: crystalline, liquid, and plastic, all of which utilize
the ionization produced by charged particles to generate optical photons, usually in the blue to green
wavelength regions. Examples of pure organic crystals are anthracene (C14H10) and trans-stilbene
(C14H12). Plastic scintillators are non-fluid solutions consisting of fluorescent organic compounds disLandolt-Brnstein
New Series VIII/4

10-12

10 Measuring techniques

[Ref. p. 10-25

solved in a solidified polymer matrix or fluid solutions with similar fluorescent organic compounds
[02Sai]. In contrast to inorganic scintillators, the fluorescence process in organic scintillators is an inherent molecular property which is characterized by the excited states of the individual molecules. Therefore,
fluorescence can be observed independent of the physical state of the material. Organic scintillators are
generally faster in their response than inorganics and are more suitable for -particle spectroscopy and
especially for fast neutron detection due to the high hydrogen fraction in their composition (cf. Section
10.1.7). Moreover, some organic scintillator materials (such as BC 501 / NE 213, cf. Tab. 10.3) offer the
possibility to discriminate between photons and neutrons due to differences in scintillator response. In
these materials, electrons released by -quanta cause scintillations at a rate faster than that due to photons
created by neutrons.
Table 10.3. Physical properties of a few representative scintillator materials (data from [02Sai]).
Material
Type
Density RefracDecay
Light
Wave- Main application
tion
outputa constantb lengthc
index
[g/cm3]
[%]
[ns]
[nm]
Inorganic scintillators
NaI(Tl)
Crystal 3.67
1.85
100
250
415
, X-rays
CsI(Tl)
Crystal 4.51
1.79
45
1005
550
, heavy particles
CsI(Na)
Crystal 4.51
1.84
85
630
420
, heavy particles
BGO
Crystal 7.13
2.15
20
300
480
, X-rays
LSO
Crystal 7.40
1.82
63
40
420
, X-rays
Organic scintillators
Anthracene
Crystal 1.25
1.62
100
30
447
General purpose
Trans-stilbene
Crystal 1.16
1.63
50
4.5
410
, fast n
BC 400 / NE 102
Plastic 1.03
1.58
65
2.4
423
General purpose
BC 422 / NE 111
Plastic 1.03
1.58
55
1.6
370
Ultra-fast timing
BC 501 / NE 213
Liquid 0.87
1.51
78
3.2
425
Fast n with discrimination
BC 509 / NE 226
Liquid 1.61
1.38
20
3.1
435
, insensitive to n
a

Given relative to NaI(Tl) for inorganic scintillators and relative to anthracene for organic scintillators. The light output of anthracene is 40-50 % of NaI(Tl). b Main component. c Maximum emission.

As mentioned above, photomultiplier tubes (PMT) have two different functions: conversion of ultraviolet and visible photons emitted by the scintillator into an electrical signal and signal amplification. Fig.
10.10 shows the essential parts of a PMT, which are mounted inside an evacuated glass envelope, namely
a photocathode, typically 10 to 12 electrodes denoted as dynodes, and an anode. The photons from the
scintillator strike the photocathode usually made of a semiconductor material formed from antimony
plus one or more alkali metals and release photoelectrons with an efficiency of about 10-30 % [94Leo].
These photoelectrons are attracted to the first dynode, which is at a higher potential than the cathode, so
that the electrons strike the dynode with a sufficiently high kinetic energy to eject three to four secondary
electrons from the surface. Since each dynode has a more positive voltage than the preceding one, this
amplification process is repeated with each successive dynode, so that a multiplication factor of 106 or
more can be obtained with a twelve stage PMT. The average gain of the dynode chain is independent of
how many electrons are simultaneously ejected from the photocathode. As a consequence, the size of the
electrical output signal at the anode is proportional to the number of electrons leaving the photocathode.
To achieve a good performance, it is important to match the emission spectrum of the scintillator to the
quantum efficiency of the photocathode material. The current measured at the anode of the PMT is fed
into an RC circuit as shown in Fig. 10.8 to produce an electrical voltage pulse.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-25]

10 Measuring techniques

10-13

The previous discussion reveals, that the output of a scintillator-PMT combination is proportional to
the amount of energy deposited by an incident particle in the scintillator and can thus be used for particle
spectroscopy. To this end, the voltage pulse goes into an amplifier and is then fed into a multichannel
analyzer, which records and stores pulses according to their voltage amplitude into different channels. By
using -ray sources of known energy, the channel numbers can be related to energy. As an example,
Fig. 10.11 shows the pulse-height spectrum of 60Co recorded from a large NaI(Tl) scintillator.
100

100
Photopeaks
Photopeaks
Backscatter

60

Compton region

40

Relative count rate [% ]

Relative count rate [% ]

80

80
Compton edges

Backscatter
3
2

1
3

20

0
0

200

400

600 800 1000 1200 1400 1600


Energy [ keV ]

200

400

600 800 1000 1200 1400 1600


Energy [ keV ]

Fig. 10.11. Pulse-height spectra of 60Co measured with (a) a large NaI(Tl) scintillation detector and (b) a highpurity germanium detector. The two characteristic photopeaks are at energies of 1.17 and 1.33 MeV. In (b) a
single (1) and a double escape peak (2) related to the incident 1.33-MeV-photons is apparent, as is an annihilation
peak at 511 keV (3) due to pair production interactions in surrounding materials. Note that incident 1.17-MeVphotons do not give rise to escape peaks, since the photon energy is only slightly above the threshold for pair
production interactions where the cross section is still very low.

A pulse-height spectrum recorded from a radiation source depends not only on the characteristics of
the radiation to be measured but also on the type of scintillator used and the mechanisms by which the
incident particles transfer their energy to the detector material. Since the ranges of charged particles are
very short in most solid and liquid materials, they deposit their energy almost completely in the detector
material giving rise to a well-defined peak in the spectrum at the particle energy. In the case of -quanta,
energy is deposited to the detector primarily by the photoelectric effect, Compton scattering, and pair
production. An incident photon undergoing a photoelectric interaction in the scintillator transfers (nearly)
all of its energy to an electron and thus contributes to the photopeak in the pulse-height spectrum, which
is located at the energy of the incident photon. In Compton scattering, however, only part of the energy is
transferred to the detector, via the recoil electrons. The scattered photon may either be absorbed by a
photoelectric interaction within the scintillator or may escape from the detector. In the first case, the total
energy of the incident photon is absorbed and the event will contribute to the photopeak. In the second
case, however, the energy deposited by the recoil electron depends on the scattering angle. The Compton
region in the spectrum thus ranges from near zero (small-angel scattering) up to a maximum energy
(Compton edge) for 180 Compton scattering. If the energy of the incident photon exceeds 1022 keV,
pair production can occur. When the positron created by this process comes to rest, it combines with an
electron to create a pair of 511 keV annihilation photons. If one or both of these photons escape, the
energy deposited in the scintillator is reduced by 511 or 1022 keV, respectively. As a consequence, additional photopeaks denoted as single and double escape peaks appear in the spectrum at energies of
511 keV and of 1022 keV below the corresponding full-energy photopeaks (cf. Fig. 10.11). Finally, lowenergy peaks may appear in the spectrum resulting from -quanta that are scattered in material outside of
the scintillator, and enter the detector having lost most of their energy. However, these backscatter peaks
Landolt-Brnstein
New Series VIII/4

10-14

10 Measuring techniques

[Ref. p. 10-25

area usually easy to identify due to their low energies (<250 keV). Since both the energy and relative
emission probability of -rays released by a radioactive decay is a characteristic property of nuclides,
-spectroscopy is widely used for the identification of nuclides in a sample or in the environment
[94ICR2]. A discussion of the various aspects related to particle spectroscopy can be found in [00Kno,
95Tso].
Scintillation detectors not only have a markedly higher detection efficiency for - or X-rays than gasfilled ionization detectors due to the higher density of the detector material but also a much shorter resolving time which allows them to respond more linearly to higher count rates than Geiger-Mller counters. Therefore, scintillation counters are frequently applied for particle counting and identification. As an
example, Fig. 10.12 shows a portable NaI spectrometer for the localization, identification, and measurement of -rays. A major limitation of scintillation detectors, on the other hand, is their poor energy
resolution. The reason is that an average energy loss of 100 eV or more in the scintillator material is required to release one photoelectron from the photocathode of the PMT and that, consequently, the average
number N of photoelectrons or information carriers produced by an incident ionizing particle is no
more than a few thousands. Due to the random nature of the interaction processes, the standard deviation
characterizing the statistical fluctuations in that number is proportional to N and the relative uncertainty proportional to 1 N . Therefore, there is a significant inherent limitation on the energy resolution of scintillation counters [00Kno]. For example, scintillation detectors used in -spectroscopy typically show an energy resolution in the range of 5 - 10 % (cf. Fig. 10.11a). The intrinsic statistical limit on
energy resolution can only be reduced by increasing the number of information carriers created per unit
energy lost by the incident radiation as, for example, by the use of semiconductor materials.

Fig. 10.12. Portable NaI spectrometer with 496 channels for the
localization, identification, and measurement of -rays with an energy
between 25 keV and 2 MeV. (Courtesy Berthold Technologies).

10.1.4 Semiconductor detectors


Semiconductor detectors have experienced a rapid development in the last decades. They are basically
solid-state analogs of gas-filled ionization detectors, in which electron-hole pairs are created by incident
ionizing radiation instead of electron-ion pairs.
Semiconductors are made of crystalline materials whose electrical conductivities are midway between
those of conductors and insulators. Their electrical properties are characterized by their crystal structure:
According to quantum theory, the energy of an electron in the crystal must fall within well-defined bands
the valence or the conduction band, which are separated by a forbidden energy gap. The most common
semiconductor materials used are silicon and germanium which crystallize in the diamond structure
(Table 10.4). In this structure, the four valence electrons form covalent bonds with each of the four
nearest neighbor atoms in the crystal and are thus at least at very low temperatures immobile. The
Landolt-Brnstein
New Series VIII/4

Ref. p. 10-25]

10 Measuring techniques

10-15

energy gap between the valence and conduction band is, however, much smaller in semiconductors (about
1 eV) than in insulators (more than 5 eV) and thus ionizing radiation, light, or heat can easily break
covalent bonds and raise electrons into the conduction band, leaving behind electron vacancies or holes
in the valence band. When an electric field is applied across a semiconductor crystal, free electrons in
the conduction band and positive holes in the valence band, which act as positive charge carriers, move
towards the positive and negative terminal, respectively, and establish a small current when the terminals
are connected to a detection circuit. The problem is, however, that at non-zero temperatures electron-hole
pairs will be thermally generated in the semiconductor material and that the random fluctuations that unavoidably occur in the resulting steady-state leakage current are too high, even in the highest resistivity
materials available, to permit the detection of the minute current caused by electron-hole pairs initiated by
an ionizing particle. This problem can be solved by using the favorable properties of a p-n semiconductor
junction, which acts as blocking contact with an extremely high resistance and thus reduces the leakage
current through the bulk of the semiconductor material.
Table 10.4. Physical properties of silicon and germanium (data from [95Tso] and [99Lut]).
Property
Silicon
Germanium
Atomic number
14
32
Atomic weight
28.1
72.6
3
2.33
5.32
Density at 300 K
[g/cm ]
1.11
0.67
Energy gap at 300 K
[eV]
500
Intrinsic resistivity at 77 K
[ m]
0.47
Intrinsic resistivity at 300 K
[ m] 2300
3.7
2.96
Average energy per electron-hole pair at 77 K [eV]
3.65
Average energy per electron-hole pair at 300 K [eV]

To this end, a pure semiconductor is doped on one side with pentavalent impurity atoms (for example,
phosphorus, antimony, or lithium) and on the other side with trivalent impurity atoms (for example, boron, gallium, or indium). When present in small concentrations, the impurity atoms will take the place of
a tetravalent normal silicon or germanium atom in the lattice and introduce only lightly bounded excess
electrons or additional electron vacancies (holes) in the crystal lattice, respectively. A semiconductor
material containing an electron-donor impurity is denoted as n-type material, material doped with a holeforming impurity as p-type material. As already mentioned, the essential part of a semiconductor detector
or diode is the region in the vicinity of the interface between the p-type and n-type material, which is
denoted as p-n junction. Due to the difference in the concentration of electrons and holes between the two
materials, electrons diffuse into the p-region and holes into the n-region. As a consequence, the diffusing
electrons fill up holes in the p-region while the diffusing holes capture electrons on the n-side, leaving a
region completely depleted of mobile charge carriers, as schematically shown in Fig. 10.13. Since p- and
n-type materials are originally electrically neutral, the diffusion process creates a net negative and
positive space charge on the p-side and n-side of the junction, respectively. At equilibrium, the electric
diffusion potential (about 0.5 - 1 V) across the p-n-junction results in the transport of charge carriers in
the opposite direction which precisely balances the diffusion process of charge carriers and limits the extension of the depletion region to rather small depths (about 50 to 100 m). However, the region where
the electric field exist and thus the depletion depth can be increased by applying a strong reverse-bias
voltage across the semiconductor, as illustrated in Fig. 10.13. It is important to note, that thermal generation of electron-hole pairs in the depletion region does not result in a considerable steady-state concentration of carriers because removal of these carriers is a much faster process than their creation. Therefore, the small concentration of carriers created by ionizing particles can easily be detected above the
highly suppressed concentration of thermally generated carriers [00Kno].

Landolt-Brnstein
New Series VIII/4

10-16

10 Measuring techniques

[Ref. p. 10-25

Pure semiconductor crystals, referred to as wafers, are grown from the melt of high-purity polycrystalline silicon or germanium material with the help of a seed crystal of defined orientation. A p-n junction
detector is usually produced by depositing sufficient p-type impurities into one end of a uniformly
n-doped wafer so as to change that end into a p-type material. This can be done either by diffusion or
implantation of dopants, or by a combination of both processes. A more detailed description of the fabrication and design of semiconductor detectors and of their application is presented in [99Lut].
Depletion
region

- -- -- -- -- - - p material

+ + + +
+

+ + +

+ + +
n material

electron
hole
negative ion

+ positive ion

Fig. 10.13. Schematic representation of a p-n detector or


diode. At the junction, electrons diffuse into the
p-region and fill up holes while holes diffuse into the
n-region and capture electrons. This process results in a
region completely depleted of mobile charge carriers (grey
area). By applying a reverse-bias voltage, the depth of the
depletion region is increased.

For particle spectroscopy, semiconductor detectors are operated in the pulse counting mode. As in the
case of a scintillation detector, the charge created in the detector material is transformed by means of a
charge sensitive preamplifier stage into a voltage pulse, the height of which is proportional to the energy
deposited by an incident ionizing particle in the depletion region. The energy resolution of a semiconductor detector, however, is much better than that of a scintillation counter, since the average energy required to produce an electron-hole pair in a semiconductor material (about 3 eV, cf. Table 10.4) is much
smaller than the average energy required to release a photoelectron from the photocathode of the PMT
(more than 100 eV). The superior energy resolution is demonstrated by the comparative pulse-height
spectra of 60Co plotted in Fig. 10.11. Whereas silicon is the most widely used semiconductor material for
charged particle detection, germanium is the preferred material for -spectroscopy because of its much
higher atomic number (cf. Table 10.4) and thus its greater photoelectric cross section. In contrast to
silicon, however, germanium must be operated at cryostatic temperatures because of its relatively narrow
energy gap between the valence and conduction band. A principal drawback associated with all semiconductor detectors is the degradation in performance which can be brought about by radiation damage.
At low energies, the efficiency of semiconductors for -detection is a function of photon cross-section
and window thickness, whereas at higher energies the total active detector volume becomes the most
important factor. Germanium detectors can be fabricated in many different geometries thus offering
devices that can be tailored to the specific needs of the measurement [cf. 95Tso]. With the exception of
the well-type configuration, the efficiency of germanium detectors is low relative to Na(Tl) scintillators.
This is, however, more than compensated for by the better energy resolution. As representative examples,
Fig. 10.14 shows detector efficiency and energy resolution curves for various types of germanium
detectors. To take full advantage of their intrinsic energy response, detectors with thin contacts such as
low-energy germanium (LEGe) and reverse electrode germanium (REGe) detectors are usually
equipped with a beryllium cryostat window.
In the last decade, compound semiconductors have gained increasing interest as detector materials
applicable to -spectroscopy [00Kno]. Particularly cadmium zinc telluride (CdZnTe) based detectors have
been developed intensively and have recently seen significant improvements [01Tak]. These devices offer
some major advantages: operation at room-temperature due to the wide band gap of the material, a resolution that is intermediate between that of scintillation detectors and germanium devices, and a high density
of the crystal providing excellent stopping power over an energy range of a few keV to over 1 MeV.
Based on these features, portable -spectrometers have been developed for radiation protection measurements.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-25]

Efficiency [%]
10

1
2
3
4
5

10-17

Resolution (FWHM) [keV ]

10

10

10 Measuring techniques

REGe, 15% rel.efficiency


LEGe, 10 cm2 15 mm thick
LEGe, 200 mm2 10 mm thick
Coaxial, 10% rel.efficiency
BEGe, 5000 mm2 30 mm thick

Well
1

Coaxial
REGe
large
LEGe
LEGe
small
LEGe

0.1

10

20

50 100 200
Energy [keV ]

500 1000 2000

5.9 10

100 122
Energy [keV ]

1322

Fig. 10.14. Typical performance parameters for various types of germanium detectors frequently used for -spectroscopy. (a) Absolute detector efficiency as a function of energy compared to that of a 37 37 mm2 Na(Tl) crystal at a
detector to source distance of 25 cm. (b) Energy resolution (full-width at half-maximum, FWHM) as a function of
photon energy. Well: well-type Ge detector, REGe: reverse electrode Ge detector, LEGe: low-energy Ge detector,
BEGe: broad energy Ge detector, Coaxial: coaxial Ge detector. (Figures adapted with permission from Canberra
Industries).

Fig. 10.15. Direct-reading electronic personal dose and


dose rate meter for measurements of X- and -rays over
the energy range from 60 keV to 3 MeV with an
accuracy of the dose reading of 15 % between 1 Sv
and 10 Sv. The detector system, which utilizes an
energy compensated silicon diode, is calibrated to give
directly the personal dose equivalent Hp(10). (Courtesy
Rados Technology).

With recent improvements in their applicability and reliability, silicon (PIN) diodes have also become
popular as radiation detectors in electronic pocket dosimeters (cf. Fig. 10.15). To measure the absorbed
dose, it is more appropriate to operate these devices in the current mode, since the current measured with
a silicon diode is nearly proportional to the absorbed dose rate in soft tissue for photon energies between
150 keV to well over 1 MeV. The reason is that the mass absorption coefficients for silicon is within
10 % of soft tissue values over the given range of photon energies and thus conversion of photons into
energetic electrons is similar in both cases [00Kno]. At lower energies, however, photon absorption in
silicon deviates considerably from that in tissue. It is thus necessary to compensate for this effect by
employing metallic filters around the detector. There are different approaches to realize energy compensation: One approach, which is designed to measure both -quanta and -particles, is to use three
diodes in parallel with individual filters to produce an appropriate energy response that is approximately
energy-independent down to about 17 keV [95Hir]. A more cost effective solution is to use a single diode
with a simple filter, usually tin, to flatten the energy response with the major disadvantage of giving up
response to photons with an energy below about 60 keV. By using a composite filter of two or more
filters together with several openings, however, it is not only possible to compensate the energy response
of a silicon diode but also to maintain an extended low energy response [96Ols]. As compared to passive
personal monitoring devices such as photographic films or thermoluminescence dosimeters, described in
the subsequent sections, electronic personal dosimeters offer various advantages, such as real-time
Landolt-Brnstein
New Series VIII/4

10-18

10 Measuring techniques

[Ref. p. 10-25

measurement and display of the dose rate and cumulative dose, audible warning of radiation fields at user
settable alarm levels, and the possibility to transmit data for remote readout and evaluation by dosimetry
management system.

10.1.5 Thermoluminescence and radiophotoluminescence detectors


The use of thermoluminescence detectors (TLDs) is another common method of solid-state dosimetry
based on the electrical properties of crystals. In these crystals, too, electrons are raised by ionizing radiation from the valence into the conduction band. In contrast to scintillation materials, however, the electrons and holes formed by the excitation process do not recombine immediately but are caught in traps
for long periods of time at room temperature, as shown in Fig. 10.16. Whereas in some natural materials
traps are formed by lattice imperfections and impurities inherent to the crystal, small concentrations of
impurity (e.g., Mn, Ti, Tm, Mg, Dy) must be added in others, which function as an activator. Some TL
materials commercially available are CaSO4:Mn, CaF2, CaF2:Mn, CaF2:Dy; Li2B4O7:Mn, LiF, LiF:Mg,
Ti, and LiF:Mg, Cu, P (activators are given after the colon). The choice among TL materials depends on
different factors: (1) the energy depth of the traps, which determines the number of charge carriers
trapped per unit of absorbed dose and thus the sensitivity of the TL detector, (2) the retention of the
trapped carriers for longer periods of time at normal temperatures, and (3) linear response over a large
dose range.
100

Electron trap

Hole trap

Incident particle

Heating

Recombination
with light
emission

Valence band

Fig. 10.16. Energy-level diagram of a TLD crystal.


Left: Radiation induced formation of an electron-hole
pair leading to the population of an electron and a hole
trap. Right: Release of a trapped electron by heat and
subsequent recombination with a hole resulting in the
emission of a photon. It is assumed that the electron is
liberated first since the depth of the electron trap is less
than that of the hole trap. In the reverse case, the hole
would be thermally released first. The dashed lines
represent drift of charge carriers in the valance and
conduction band.

Relative thermoluminescence intensity [%]

Excitation by
radiation

Conduction band

LiF:Mg, Ti
CaF2 :Dy
80

60

40

20

10
Time [s ]

15

20

Fig. 10.17. Representative thermoluminescence glow


curves of LiF:Mg, Ti (TLD-100) and CaF2:Dy heated
from 50 C to 325 C within 20 s. The curves are
normalized to the same maximum intensity.

The charge carriers stored in a TLD after having been exposed to radiation can be released by heating
the detector, e.g., by a stream of heated gas, by laser light, or by heating the support. As schematically
shown in Fig. 10.16, this process provides sufficient thermal energy to electrons or holes so that they are
raised back into the valence or conduction band, respectively. Free electrons (or holes) can migrate
towards the position of a trapped hole (or electron) and recombine with the emission of light. The photons
emitted by the heating process are detected by a photomultiplier tube (PMT) as in the case of a scintillation detector (cf. Section 10.1.3). The PMT output signal, which is proportional to the number of photons
released in the TL material, is detected and plotted as a function of temperature or time during heating
Landolt-Brnstein
New Series VIII/4

Ref. p. 10-25]

10 Measuring techniques

10-19

with a constant rate. The resulting plot is denoted as glow curve, which may consist of various resolved
or unresolved glow peaks if there are several types of traps in the TLD material each of which is
characterized by a specific depth or binding energy (Fig. 10.17). The area under either the entire curve or
individual peaks is a measure of the number of trapped charge carriers and thus of the amount of energy
absorbed in the crystal. Determination and analysis of glow curves are performed by dedicated devices
denoted as TLD readers (Fig. 10.18). Since the intrinsic TL efficiency, i.e. the ratio between the light
energy emitted per unit of detector mass and the absorbed dose, is in the order of 1 % only, TLDs must be
used under reproducible conditions to obtain consistent results. One important aspect is to thoroughly
deplete all traps in the detector before it is reused for dose measurements. To this end, the temperature of
TLDs must be raised to relatively high values over longer periods of time in an annealing oven (cf. Fig.
10.18). TLD dosimetry is a relative measurement technique and thus TLDs have to be calibrated
individually against absolute dosimetry systems such as a calibrated ion chamber. Details on TLD
dosimetry and calibration are presented in [86Att, 00Sha].
TLDs are available in diverse forms such as loose powder, chips, rods, rings, or small wafers and
are widely used as single element dosimeters or assemblies for a large variety of radiation protection
measurements (Fig. 10.18). They have found, for example, an important place in personal monitoring and
are rapidly replacing the use of photographic films discussed in the next section. In personal monitoring
devices, one or more small TLD elements are usually assembled into rigid aluminum cards and mounted
within shielded filter-holders. For this and many other purposes, LiF:Mg, Ti is the material of choice
because it is approximately tissue-equivalent (effective atomic number of 8.2 compared to 7.4 for tissue)
and almost energy-independent for photons with an energy between 0.1 and 3 MeV [86Att]. Moreover, it
is to some extent sensitive to thermal neutrons, since natural lithium (TLD-100) contains to 7.4 % 6Li that
has a high (n,) capture cross section. The response to neutrons can be enhanced or reduced by using 6Li
(TLD-600, with 96.5 % 6Li) or 7Li enriched material (TLD-700, with 99.99 % 7Li) (cf. Section 10.1.7).
a

Fig. 10.18. (a) TL detectors for radiation protection measurements (chips, rods, and ring). (b) TLD reader for the
evaluation of irradiated detectors. (c) Annealing oven for the preparation of the detectors. TL detectors are placed
in a metallic container which comes into contact with two heating plates to ensure optimal thermal equilibrium.
(Courtesy PTW Freiburg).

In some substances such as BeO, LiF, Al2O3, CaSO4, and some alkali halides electrons liberated
during the heating procedure may also have a chance to leave the material when the trap sites are located
in a thin (<10 nm) surface layer. This process, that is closely related with the conventional TL mechanism
[99Sak], is referred to as thermally stimulated exoelectron emission (TSEE). The low energy exoelectrons
can be detected, for example, by a windowless proportional or Geiger-Mller counter (cf. Section
10.1.2.3 and 10.1.2.4, respectively). The number of exoelectrons detected is proportional to the dose in
the surface layer. TSEE dosimetry has gained interest since it can be used for the measurement of weakly
penetrating radiation, such as low energy -quanta or - and -particles [86Sch].
A limitation of conventional TLD technology is the complete annealing of populated traps, when the
material is heated during the readout process. Therefore, alternative luminescence techniques have been
investigated that permit successive readouts and the construction of integrating dosimeters. An approach
that has been applied more frequently in health physics is based on the optical phenomenon of radiophotoluminescence (RPL) [68Att, 87Per]. RPL is a property of certain substances to emit fluorescent light
Landolt-Brnstein
New Series VIII/4

10-20

10 Measuring techniques

[Ref. p. 10-25

in the visible range upon irradiation with ultraviolet light, when the substance has previously been
exposed to ionizing radiation. Although various materials exhibit this property, silver-activated metaphosphate glasses are most commonly used. In RPL glasses, ionizing radiation results in the formation of
stable fluorescing centers that emit orange light (about 640 nm) under (pulsed laser) ultraviolet stimulation (365 nm) [93Pie]. The intensity of fluorescence light, which is determined with a spectrally matched
PMT as in the case of traditional TLD readout, is proportional to the number of light centers and hence to
the radiation dose. Since the light centers are not destroyed by ultraviolet excitation, the readout procedure can be repeated as often as necessary. For practical application as personal dosimeters, optimized
RPL (Yokato) glasses are covered with metal filters or capsules which provide largely energy-independent dose measurements for -quanta with an energy above 50 keV [72Kie, 96Hoh].

10.1.6 Photographic films


Film dosimetry is an attractive technique for many applications especially in medical physics due to
its high spatial resolution, wide accessibility, and the flexibility to place the film in humanoid phantoms
[00Sha]. Furthermore, photographic films are still widely used for monitoring radiation exposure of personnel although they do by no means meet the requirements of an ideal personal dosimeter in all respects.
Photographic films consist of an emulsion of microscopic grains of silver halides, usually silver bromide,
dispersed in a gelatine layer on one or both sides of a transparent film base of cellulose acetate or
polyester. The photographic process is very complicated. A thorough discussion of the complex experimental and theoretical aspects, which are considerably simplified in the following description, can be
found in [02Bus] and [81Bar], respectively. When the silver bromide grains in the emulsion are exposed
to visible light or ionizing radiation, bromide ions absorb energy and are oxidized. The electrons from this
oxidation process reduces silver ions to silver atoms. Experimental evidence indicates that a minimum of
three to five reduced silver atoms form a sensitized grain, that can act as a catalyst for the chemical
amplification process during film development. When the film is placed in a chemical developer solution,
all silver ions in the grains will be reduced to silver atoms independently of whether the grains were
affected by ionizing radiation or not. Silver atoms in sensitized grains, however, greatly enhance the rate
of reduction of additional silver ions. Based on this fact, the chemical development process is terminated
after a time at which the reduction process in sensitized grains is completed by taking the film out of the
developer solution and placing it in a fixing solution that neutralizes residual developer present on the
film. The chemical development process greatly increases the number of silver atoms and makes the
radiation effect measurable. To ensure reproducibility of the results, careful control of the developing
procedures is essential.
The degree of macroscopic blackening of the processed film depends on the number of silver atoms
deposited and thus on the amount of energy absorbed. It is usually expressed in terms of the optical density OD which is defined in terms of the transmission of light through the film as OD = log 10 ( I 0 I ) ,
where I 0 and I are the intensities of a light beam measured by an optical densitometer in front and behind
the film. The relation between the optical density of the film and the exposure is described by the characteristic curve for the particular film typically a sigmoid-shaped curve with three characteristic regions: At low exposures, the optical density is low and independent of exposure level. Next, there is a
segment over which the optical density and the logarithm of the exposure are approximately linear related. This is the region of normal operation. In the third segment, which corresponds to large exposures,
the film becomes saturated since all silver ions are converted to metallic silver [81Bar].
The response of photographic films is strongly dependent on radiation energy. For photon energies
below 100 keV, for example, the relative sensitivity is between 10 and 50 times higher than at higher energies [86Att]. In order to flatten the energy response, film badge dosimeters contain a set of metallic
filters of various materials and thicknesses over different regions of the film as shown in Fig. 10.19. By
comparison of the optical density behind these filters, it is possible to get rough spectral information
Landolt-Brnstein
New Series VIII/4

Ref. p. 10-25]

10 Measuring techniques

10-21

which can be utilized to take the energy dependence of the films sensitivity approximately into account.
Disadvantages of film badge dosimeters are, firstly, the considerable angular dependence of the detection
efficiency and, secondly, fading of sensitized grains in the period between exposure and development
[72Kie]. Both effects can result in a considerable underestimation of radiation exposure.
Fig. 10.19. Film-badge dosimeter
for personal monitoring. The interior view of the holder shows at
the back and front an open
window, three copper filters of
different thickness, and a staggered
lead / tungsten filter to identify the
direction of the incident radiation.
The light-tight wrapping contains
two dosimetry films of different
sensitivities.

10.1.7 Detectors for neutrons


In the previous sections, devices for the detection of photons and charged particles have been described,
which deposit their energy in matter predominantly via electromagnetic interactions in particular by
inelastic collisions with the atomic electrons. In contrast, the interaction of fast neutrons with matter
occurs through processes with the nucleus, e.g. elastic or inelastic scattering or various nuclear reactions.
The most important nuclear reactions for the detection of neutrons in the eV to keV region are (n,) and
(n,p) reactions, such as 6Li (n,) 3H, 10B (n,) 7Li, or 3He (n,p) 3H, in which a neutron is captured and a
charged particle is released. These substances have a high sensitivity at low neutron energies, which is of
particular advantage considering the low dose rate levels routinely encountered in radiation protection,
but become more and more ineffective at higher neutron energies E because the capture cross sections
vary as 1 / E . Instead, elastic scattering of neutrons at nuclei becomes the most relevant interaction
mechanisms, giving rise to recoil nuclei. Whereas the energy transfer to a recoil nucleus is very ineffective in the case of heavy nuclei, an incident neutron can transfer up to its entire energy in a single collision to a hydrogen nuclei (proton) because neutrons and protons have nearly the same mass. Therefore,
detectors with a high content of hydogen in the detector or cover material are used for neutron detection
by measuring the recoil protons. In addition, hydrogen-containing materials, so-called moderators, can be
utilized for slowing down of fast neutrons to thermal energies. A more quantitative discussion of the
various interaction processes, by which neutrons transfer all or part of their energy to charged particles
capable of exciting and ionizing, can be found in [72Kie, 00Kno, 94Leo].
A sensitive and simple counting device for slow neutrons is a proportional counter filled with BF3 gas,
usually enriched to more than 90 % in 10B. In such a counter, BF3 gas not only serves as the target for
slow neutron conversion into -particles via the above mentioned 10B (n,) 7Li reaction but also as filling
gas. Since -particles yield a much higher output signal than -particles, BF3 counters can be used to
discriminate between the neutron and -component in a mixed n- field (cf. Section 10.1.2.3). By sourrounding the counter by a moderating material (e.g. paraffin or polyethylene) in which the neutrons are
slowed down, it can also be utilized for the detection of fast neutrons. An alternative approach for the
detection of slow neutrons is the use of proportional counter, the walls of which are coated with a layer of
B2O3 or BC4. Such boron-lined counters offer the flexibility to use more appropriate filling gases than
BF3 [00Kno, 95Tso].
A particular problem in neutron dosimetry is the broad energy range that can occur and the considerable variation of detector response over this range. Nevertheless, dosimeters for routine area monitoring
can be constructed in such a way that the shape of their fluence response as a function of energy approximate that of the fluence-to-dose equivalent conversion function and thus will give a reading approximately proportional to the ambient dose equivalent H*(10) over most of the energy range of interest
Landolt-Brnstein
New Series VIII/4

10-22

10 Measuring techniques

[Ref. p. 10-25

(cf. Section 4.5.3.3 and [01ICR]). To this end, thermal-neutron detectors are surrounded by a moderator,
whose material and geometry is chosen to optimize the desired response function. A widely used survey
instrument is the rem counter shown in Fig. 10.20. It consists of a massive sphere with a diameter
between 25 and 30 cm of hydrogenous material usually polyethylene with additional layers of cadmium
or boron that serves as neutron moderator surrounding a thermal-neutron detector, e.g. a 3He
proportional tube. An extension of this approach is the Bonner sphere spectrometer, which uses
different moderator spheres with diameters ranging from 5 to 30 cm. Since the response functions for the
diverse moderator spheres differ in shape and position of the maxima, the energy distribution of a neutron
field can be derived from count rate measurements performed separately for each of the spheres [00Kno,
00Sha].
Shielding
Front window
6
7

Body

LiF TLD
LiF TLD

Albedo window
Shielding

Fig. 10.20. Dose rate monitor (rem counter) for the


measurement of neutron ambient dose equivalent
H*(10) for neutrons up to an energy of 20 MeV. The
detector system consists of a proportional counter tube
centered in a moderator sphere with a diameter of 250
mm. (Courtesy Berthold Technologies).

Fig. 10.21. Schematic representation of an albedo TLD


dosimeter consisting of two pairs of 6LiF - 7LiF detectors
that are shielded by cadmium- or boron-loaded material
either against incident thermal neutrons or albedo
neutrons from the rear.

As described in Section 10.1.2.3, tissue-equivalent proportional counters (TEPCs) are capable of


measuring absorbed dose to the sensitive gas volume and of determining an approximation of the dose
equivalent by its spectroscopic properties. They thus provide additional information normally not available from other neutron-measuring instruments. Due to practical limitations, however, TEPCs are not yet
readily usable in everyday routine monitoring [01ICR, 00Sha].
For personal monitoring in mixed n- fields, the most commonly used devices today are albedo TLD
badges, containing TL detector chips with a high fraction of 6LiF (such as TLD-600), that has a large
6
Li (n,) 3H cross section for thermal neutrons, as well as chips with a high fraction of 7LiF (such as
TLD-700), that is insensitive to neutrons. The dosimetry of fast neutrons becomes possible when the
batch is held closely to the body where it will be exposed to low energy neutrons that are backscattered
from the body as the result of the moderation of fast neutrons within the body. These scattered neutrons
are called albedo neutrons. As an example, Fig. 10.21 shows the schematic design of a TLD albedo dosimeter that consists of two pairs of 6LiF - 7LiF detectors. One pair is shielded from the rear with a thermal-neutron absorber (e.g. cadmium- or boron-loaded plastic) the second from the front. The readings
obtained from the incident-neutron and albedo-neutron detectors are combined into an overall calculation
of neutron dose equivalent. It should be mentioned, however that the resulting dose equivalent response
of the dosimeter varies greatly with neutron energy at intermediate and high energies. Therefore, a single
calibration factor cannot be used in different neutron fields with widely varying spectra if accurate dose
results are to be obtained. In such cases, it is necessary to keep a record of the location in which the TLD
batch was used, and to apply the appropriate calibration factor for the reading [01ICR].
Landolt-Brnstein
New Series VIII/4

Ref. p. 10-25]

10 Measuring techniques

10-23

Alternatively, etched-track detectors are used for neutron personal dosimetry. These passive devices
utilize the ability of some materials to register the tracks of (neutron induced) charged particles as damage
to their structure. The permanent damage tracks can be greatly enlarged by chemical or electrochemical
etching procedures. The resulting conical etched pits in the material at the damage sites are visible to the
naked eye and can be counted automatically by optical systems. Most frequently, plastic foils of
polyallyl diglycol carbonate (PADC, also known by the trade name CR-39) are used, which are
uniquely sensitive to recoil protons released by neutrons passing through the material. PADC is sensitive
to fast neutrons with an energy down to about 100 keV, and, by adding a converter layer, also to thermal
and epithermal neutrons. PADS foils are insensitive to photons and -particles and are thus suitable to
measure the neutron component in a mixed n- field [01ICR, 97Bar]. (Due to the same reason, PADC
detectors are used to estimate personal radon doses as well as integrated indoor radon concentrations by
detecting the -particles from the decays of radon and its daughter products.)
Superheated-drop or bubble detectors can be employed for both area and personal monitoring. They
consist of microscopic droplets of liquid halocarbons and hydrocarbons dispersed in a gel or polymer
matrix. The liquid droplets are superheated and are thus in a metastable state. When a neutron interacts
with a nucleus inside or near a droplet, the resulting secondary charged particles locally increase the
temperature and thus cause local vaporization. By this process, small vapor bubbles are formed that begin
to expand by vaporizing adjoining liquid. If a vapor bubble reaches a critical size, all of the liquid in the
droplet will be vaporized resulting in a visible gas bubble with a diameter of up to a millimeter. If the
neutron energy exceeds a specific detector threshold which depends on the atomic composition and size
of the superheated droplets and the temperature and pressure of the matrix the overall absorbed energy
is correlated to the number of bubbles in a vial [01ICR, 00Kno, 00Sha]. Typical sensitivities are in the
range of a few bubbles per Sv. Bubble detectors can be constructed as passive or active devices. In the
first case, the superheated droplets are dispersed in a viscous polymer gel so that the bubbles remain fixed
and thus can be counted at the end of the measurement by eye or automatically using an optical scanner.
Active devices can be realized, for example, by placing an electro-acoustic transducer in contact with the
detector, so that each time a bubble is formed, the sound that is produced is converted into an electrical
pulse. An important feature of all superheated drop detectors is their almost complete insensitivity to
photons and electrons with energies up to about 6 MeV [01ICR].
Further types of neutron detectors for area and personal monitoring as well as guidance concerning the
measurement of operational dose equivalent quantities for neutron radiation are given in [01ICR, 99Alb].

10.1.8 Biological dosimetry


The human genome contained in the cell nucleus is physically carried by 46 chromosomes, each of which
is composed of an extremely long, double-stranded helical DNA molecule in a closely packed form.
Chromosomes are clearly visible through a light microscope during the metaphase a certain stage of cell
division, in which the condensed chromosomes become attached to spindle fibers and lie in a central
plane of the nucleus. In this stage of cell cycle, chromosomes are already replicated (doubled). The newly
formed twin chromosomes, which are called chromatides, remain temporarily attached to each other at a
point, the centromere, located near the center of each of the chromatides (Fig. 10.22a).
There is a large body of evidence that biological effects of ionizing radiation result principally from
damage to DNA. The most important effects caused by ionizing radiation are breaks in the DNA double
helix. Single-strand breaks are restored immediately by molecular repair mechanisms using the opposite
DNA strand as a template. If the repair is incorrect, the genetic information is altered. Although such
point mutations may lead to inherited effects of offspring or the induction of carcinogenesis, they do not
result in chromosomal aberrations visible under the microscope. On the other hand, breaks at both strands
on opposite sides of the double helix that are juxtaposed may lead to a double-strand break. When this
happens, different types of chromosomal aberrations can be observed: If the radiation-induced damage
occurs before the chromosomes have been replicated, the lesion is dublicated during DNA synthesis and
thus both chromatides show the same aberration (chromosome aberration). If, on the other hand, the cell
Landolt-Brnstein
New Series VIII/4

10-24

10 Measuring techniques

[Ref. p. 10-25

is irradiated after the DNA material has already been doubled, only one of the two twin chromatides is
damaged (chromatide aberration). Gross distortions that are clearly visible in the metaphase are di- and
tricentric chromosomes. They are formed when a break occurs in two or even three chromosomes and the
centric fragments carrying the centromere incorrectly join with each other at their broken ends. The
remaining acentric fragments are lacking a centromere and can thus easily be detected in the metaphase,
too. A detailed description of the various types of radiation-induced chromosomal aberrations is presented
in [00Hal].
Cytogenetic analysis of chromosomal aberrations is a valuable tool for retrospective dose assessment
of individuals that are accidentally overexposed to ionizing radiation. It fills a gap in dosimetry, since the
radiation effect on the human body is determined directly without the intermediary of dosimetric measurements using technical devices. Scoring of chromosome aberrations in peripheral blood lymphocytes,
mainly the dicentric assay, is regarded as the most specific method and has become a routine component
of accidental dose assessment [02Voi]. Figure 10.22a shows representative chromosome aberrations in a
metaphase preparation of irradiated lymphocytes. Many studies in animals and humans have shown a
good relation between the results obtained in vivo and in vitro and this provides evidence that in vitro established dose-effect relationships can be used as calibration curves [02Voi]. However, as shown in
Fig. 10.22b, the formation of dicentrics strongly depends on radiation quality and dose rate so that
information about these variables needs to be established for each investigation. As mentioned above,
DNA breaks must be induced in two different unreplicated chromosomes in order to form a dicentric
chromosome. Since this can be achieved either by a single particle breaking on occasion both
chromosomes or two particles each of which damaging only one chromosome, the frequency N of dicentric chromosomes per cell can be well fitted as a function of dose D by a linear-quadratic model,
N = D + D2. At low doses, one-particle events (described by the linear term, D) are more frequent,
whereas two-particle events (described by the quadratic term, D2) dominate at high doses. Besides the
calibration curve used, the precision of dose estimation depends mainly on the number of cells observed
and the background level. In practice, detection of low LET radiation is possible for doses above 150
mGy [02Voi].
0.8

b
Dicentric chromosomes per cell

0.6

-rays, 60Co, 0.1 Gy/min


X-rays, 250 kVp, 1.0 Gy/min
X-rays, 90 kVp, 0.1 Gy/min
neutrons, 15 MeV
neutrons, 0.7 MeV

0.4

0.2

0.5

1.0
Dose [Gy ]

1.5

2.0

Fig. 10.22. Radiation-induced chromosomal aberrations in human lymphocytes. (a) Metaphase preparation with
normal, dicentric (D), and tricentric chromosomes (T) as well as various fragments (F). (b) Frequency of dicentric
chromosome aberrations per cell for several types of radiation. The curves give the result of linear (low-energy
neutrons) and linear-quadratic fits to measured data. (Courtesy G. Stephan, BfS).

Landolt-Brnstein
New Series VIII/4

10 Measuring techniques

10-25

10.1.9 References for 10.1


68Att
72Kie
79ICR
81Bar
83ICR
86Att
86Sch
87Per
89Die
89Men
93Pie
94ICR1
94ICR2
94Leo
95Hir
95ISO
95Sch
95Tso
95Wak
96Hoh
96Kah
96Ols
97Bar
99Alb

99Lut
99Sak
99Wer
00Hal
00Kno
00Sha

Attix, H., Roesch, W.C., Tochilin, E.: Radiation Dosimetry. 2nd edition, Vols. I-III. New York:
Academic Press, 1968-1969.
Kiefer, H., Maushardt, R.: Radiation Protection Measurement. Oxford: Pergamon Press, 1972.
ICRU Report 31: Average energy required to produce an ion pair. Bethesda, MD: ICRU
Publications, 1979.
Barrett, H.H., Swindell, W.: Radiological Imaging. London: Academic Press, 1981.
ICRU Report 36: Microdosimetry. Bethesda, MD: ICRU Publications, 1983.
Attix, F.H.: Introduction to radiological physics and radiation dosimetry. New York: John
Wiley & Sons Inc., 1986.
Scharmann, A, Kriegseis, W.: Radiat. Prot. Dosim. 17 (1986) 359.
Perry, J.A.: RPL Dosimetry: Radiophotoluminescence in health physics. Bristol: IOP
Publishing, 1987.
Dietze, G., Menzel, H.G., Schuhmacher, H.: Radiat. Prot. Dosim. 28 (1989) 77.
Menzel, H.G., Paretzke, H.G., Booz, J. (eds.): Implementation of dose-equivalent meters based
on microdosimetric techniques in radiation protection; Radiat. Prot. Dosim. 29 (1-2) (1989).
Piesch, E., Burgkhardt, B., Vilgis, M.: Radiat. Prot. Dosim. 47 (1993) 409.
ICRU Report 52: Particle counting in radioactivity measurements. Bethesda, MD: ICRU
Publications, 1994.
ICRU Report 53: Gamma-ray spectrometry in the environment. Bethesda, MD: ICRU
Publications, 1994.
Leo, W.R.: Techniques for nuclear and particle physics experiments, 2nd edition. Berlin:
Springer-Verlag, 1994.
Hirning, C.R.; Yuen, P.S.: Health Phys. 69 (1995) 46.
International Organization for Standardization: Guide to the expression of uncertainty in
measurement. Geneva, Switzerland: ISO, 1995 (corrected of first print in 1993).
Schmitz, Th., Waker, A.J., Kliauga, P., Zoetelief, H. (eds.): Design, construction and use of
tissue-equivalent proportional counters EURADOS report; Radiat. Prot. Dosim. 61 (4)
(1995)
Tsoulfanidis, N.: Measurement and detection of radiation, 2nd edition. Washington: Taylor &
Francis, 1995.
Waker, A.J.: Radiat. Prot. Dosim. 61 (1995) 297.
Hohlfeld. K.: Nachweismethoden fr ionisierende Strahlung, in: Kose, V.; Wagner, S. (eds.),
Praktische Physik, Band 2. Stuttgart: B.G. Teubner, 1996, Kapitel 7.4.
Kahilainen, J.: Radiat. Prot. Dosim. 66 (1996) 459.
Olsher, R.H., Eisen, Y.: Radiat. Prot. Dosim. 67 (1996) 271.
Bartlett, D.T.; Steele, J.D., Tanner, R.J., Gilvin, P.J., Shaw, P.V., Lavelle, J.: Radiat. Prot.
Dosim. 70 (1997) 161.
Alberts, W.G., Arend, E., Barelaud, B., Curzio, G., Decossas, J.L., drrico, F., Fiechtner, A.,
Grillmaier, R., Meulders, J.-P., Menard, S., Roos, H., Schuhmacher, H., Thevenin, J.-C.,
Wernli, C., Wimmer, S.: Advanced methods of active neutron dosimetry for individual
monitoring and radiation field analysis (ANDO), Report PTB-N-39, Braunschweig, 1999.
Lutz, G.: Semiconductor radiation detectors. Device Physics. Berlin: Springer-Verlag, 1999.
Sakurai, T.; Tomita, A., Fukuda, Y.: J. Phys. D 32 (1999) 2290.
Wernli, C., Fiechtner, A., Kahilainen, J.: Radiat. Prot. Dosim. 84 (1999) 331.
Hall, E.J.: Radiobiology for the radiologist, 5th edition. Philadelphia: Lippincott Williams &
Wilkins, 2000.
Knoll, G.F.: Radiation detection and measurement, 3rd edition. New York: John Wiley & Sons,
Inc. 2000.
Shani, G.: Radiation dosimetry: Instrumentation and methods, 2nd edition. Boca Raton: CRC
Press 2000.

Landolt-Brnstein
New Series VIII/4

10-26
01ICR
01Tak
02Bus
02ISO
02Sai
02Voi
02Wak

10 Measuring techniques
ICRU Report 66: Determination of operational dose equivalent quantities for neutrons. Journal
of the ICRU, Volume 1, No 3, 2001.
Takahashi, T; Watanabe, S.: IEEE Trans. Nucl. Sci. 48 (2001) 950.
Bushberg, J.T.; Seibert, J.A., Leidholdt, E.M., Boone, J.M.: The Essential Physics of Medical
Imaging, 2nd edition. Philadelphia: Lippincott Williams & Wilkins, 2002,.
ISO/ASTM51707-2002(E): Guide for estimating uncertainties in dosimetry for radiation
processing. Geneva, Switzerland: International Organisation for Standardization, 2002.
Saint-Gobain Crystals & Detectors: Product Data Sheets, www.detectors.saint-gobain.com.
Voisin, P.; Barquinero, F., Blakely, B., Lindholm, C., Lloyd, D., Luccioni, C., Miller, S.,
Pallitti, F., Prasanna, P.G., Stephan, G., Thierens, H., Turai, I., Wilkinson, D., Wojcik, A.:
Cell. Mol. Biol. 48 (2002) 501.
Waker, A.J.; Schrewe, U., Burmeister, J., Dubeau, J., Surette, R.A.: Radiat. Prot. Dosim. 99
(2002) 311.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-39]

10 Measuring techniques

10-27

10.2 Radiological protection measurements: external exposure


Radiological protection aims at the restriction of the doses to the human body, the effective dose and the
equivalent dose in an organ or tissue by applying constraints and limits. The assessment of these doses is
therefore fundamental to the practice of radiological protection. However, neither the equivalent dose in
an organ or tissue nor the effective dose can be measured directly. Values of these quantities must be
inferred from measurable quantities with the aid of models. Radiological protection measurements
therefore include measurements related to the system of radiological protection and the interpretation of
these measurements in the assessment of external and internal exposures. For monitoring external
exposure specific operational dose quantities have been defined which normally provide an estimate of
effective dose sufficiently accurate for the purpose of radiological protection (see Sect. 10.2.1). Details
about the types of detectors, which can be used to measure these operational quantities are described in
Sect. 10.1.
The primary justification of any monitoring program is to achieve and demonstrate an appropriate
level of protection. Further objectives of monitoring programs are to
estimate the actual radiation exposure level,
demonstrate compliance with legal requirements,
demonstrate good working practices,
provide data for use in reviewing optimization programs,
provide data for medical purposes as required,
provide data for use in epidemiological studies.
Monitoring programs can be distinguished with regard to the objectives and the location of
monitoring. While area monitoring provides a dose or dose rate which enable to estimate the dose a
person would receive when staying for a specified time period at the location of interest, individual
monitoring provides an estimate of the dose a person has already received. Monitoring can be organized
as routine, task-related and special monitoring. Local monitoring can be performed at the workplace, e.g.
by means of area monitoring. Individual monitoring can be performed by measuring the external
exposure, the internal exposure, and the skin contamination.
Routine monitoring is associated with continuous operation and is largely of confirmatory nature.
Operational individual monitoring is associated with a particular operation. It may make use of
supplementary dosimeters in addition to those used for routine monitoring. Special individual monitoring
will be applied in actual or suspected abnormal conditions including incidents and accidents.
The result of monitoring may be used to initiate certain actions when a pre-defined dose level is
exceeded.

10.2.1 Operational quantities


The International Commission on Radiation Units and Measurements (ICRU) has defined a set of
operational dose quantities for area and individual monitoring of external exposure [85ICR, 92ICR,
93ICR] which were designed to provide an estimate of the protection quantities defined by ICRP [77ICR]
and to serve as calibration quantities for dosimeters used in monitoring. More information about the
definition of the operational quantities is given in Sect. 4.5.
For area monitoring, the appropriate operational quantities are the ambient dose equivalent H*(10) for
strongly penetrating radiation, and the directional dose equivalent H'(0.07, ), for weakly penetrating
radiation (see Sect. 4.5.3.3).
For individual monitoring, the quantity personal dose equivalent Hp(d ) was defined, which is the dose
equivalent in ICRU soft tissue, at an appropriate depth d below a specified point on the body where the
individual dosimeter is worn [92ICR, 93ICR]. For strongly penetrating radiation a depth of 10 mm,
denoted by Hp(10), and for weakly penetrating radiation a depth of 0.07 mm, denoted by Hp(0.07), is
used. A depth of 3 mm, denoted by Hp(3), was also proposed for monitoring the exposure of the lens of
the eyes but has never been used in practice.

Landolt-Brnstein
New Series VIII/4

10-28

10 Measuring techniques

[Ref. p. 10-39

Personal dose equivalent is defined in the human body and may, therefore, in a given exposure
situation vary between individuals. The value may also depend on the position of the dosimeter worn on
the body. Consequently, the personal dose equivalent can be expected to vary between locations of any
given individual and is anticipated to be a multi-valued quantity [96ICR, 98ICR, 99Zan]. For routine
monitoring in cases where the readings are far beyond the corresponding legal limits those values are seen
to provide a sufficient approximation to the corresponding protection quantity, e. g. effective dose, if the
dosimeter is worn at a position representative for the exposure. To make this quantity single-valued in a
given exposure situation, both a particular location of the dosimeter on the human body and a particular
phantom of the body need to be specified for evaluation. More information is given in Chap. 6.
Table 10.5 summarizes the objective of dose control and the corresponding operational dose quantities
used and specifies their application.
Table 10.5. Operational dose quantities and their objectives in external monitoring
Dose quantities for
Objective
area monitoring
individual monitoring
control of effective dose
ambient dose equivalent, H*(10) personal dose equivalent, Hp(10)
control of skin equivalent dose directional dose equivalent,
personal dose equivalent, Hp(0.07)
H'(0.07)
control of equivalent dose of
directional dose equivalent, H'(3) personal dose equivalent, Hp(3)
the eye lens
The operational quantities for area and individual monitoring of external exposure are chosen to
approximately assess the effective dose under most exposure conditions. Because of the different models
used in the definition of the quantities the ratio of the operational quantities and the effective dose
depends on the type and the energy of the radiation considered and on the direction of radiation incidence
on the body. Figs. 10.23, 10.24, and 10.25 show the ratios E/H*(10) and E/Hp(10) for photons and
neutrons under various exposure conditions.
1.6

E / H * (10)

1.2
1.0

AP
PA
RLAT
LLAT
ROT

E / Hp (10)

1.4

AP
PA
RLAT
LLAT
ROT
ISO

0.8
0.6

0.4
1

0.2
0
10 2

1
10-1
Photon energy [ MeV ]

10

Fig. 10.23. Ratio of effective dose E and ambient dose


equivalent H*(10) versus photon energy for various
directions of photon radiation incidence on the human
body [96ICR]. AP: frontal incidence, LLAT: left lateral
incidence, RLAT: right lateral incidence, PA: incidence
from the back, ROT: incidence rotational to the vertical
axis, ISO: isotropic incidence.

0
10-2

1
10-1
Photon energy [ MeV ]

10

Fig. 10.24. Ratio of effective dose E and personal dose


equivalent Hp(10) versus photon energy for various
directions of photon radiation incidence on the human
body and the dosimeter worn in front of the lung
[96ICR, 99Zan]. AP: frontal incidence, LLAT: left
lateral incidence, RLAT: right lateral incidence, PA:
incidence from the back, ROT: incidence rotational to
the vertical axis.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-39]

10 Measuring techniques

10-29

2.5

E / H * (10)

2.0

1.5

AP
PA
RLAT
LLAT
ROT
ISO

1.0

0.5

0
10 9 10-8 10 7 10 6 10 5 10 4 10-3 10 2 10 1 1
Neutron energy [ MeV ]

10 10 2

Fig. 10.25. Ratio of effective dose E and ambient dose


equivalent H*(10) versus neutron energy for various
directions of neutron radiation incidence on the human
body [96ICR]. AP: frontal incidence, LLAT: left lateral
incidence, RLAT: right lateral incidence, PA: incidence
from the back, ROT: incidence rotational to the vertical
axis, ISO: isotropic incidence.

10.2.2 Reference levels


Reference levels are values of measured quantities above which specific actions or decisions should be
taken. In the context of this section the most important reference levels are the Investigation Level and
the Intervention Level also called Action Level (see Sect. 4.8). In practical implementation of
monitoring programs additional reference levels might be required. They may include levels for recording
the monitoring results (Recording Level) and for their reporting (Reporting Level or Notification
Level).
Measured values above the Investigation Level require an investigation of the reason and the
implication of the measured value. Investigation Levels are specifically defined by the operating
management and they can apply both to the individual and the working environment. It is appropriate to
select Investigation Levels for individual dose and intake on the basis of expected levels or on the basis
of a selected fraction of the relevant dose limit. Investigation Levels should be defined at the planning
stage of any practice although they may need to be revised on the basis of operational experience.
In the medical field specific Diagnostic Reference Levels for patients and for standard applications
of ionizing radiation and radioactive substances are defined which characterize a dose level corresponding
to the technical and operational state of the art which should be considered for avoiding situations where
the level of dose to a patient or the administered activity is unusually high.
Intervention applies to those situations where the source, pathways and exposed individuals are
already in place when the decisions about control or remedial measures are being considered.
Intervention Levels are set by competent authorities and are often mandatory. Typical examples of
interventions are actions taken after a radiological emergency to protect the members of the public. There
may also be the need to undertake an intervention to protect workers involved in accidents at the
workplace. Intervention may also be necessary to decrease the exposure of workers in de-facto situations,
e.g. to elevated levels of natural radiation.

10.2.3 Types of exposure


For the purpose of discussing the various aspects of radiological protection measurements it is convenient
to distinguish between occupational and public exposure.
Landolt-Brnstein
New Series VIII/4

10-30

10 Measuring techniques

[Ref. p. 10-39

10.2.3.1 Occupational exposure


Radiation work is defined [91ICR1, 96EU] as work in which the annual effective dose of an exposed
worker of age 18 or over from radiation sources at work may exceed the annual dose limits for members
of the public, e.g. an effective dose of 1 mSv or an equivalent dose of the lens of the eye of 15 mSv or an
equivalent dose of the skin of 50 mSv averaged over any 1 cm2 area, regardless of the area exposed. For
occupational exposure the ICRP has recommended a limit on the effective dose of 20 mSv per year
averaged over 5 years (100 mSv in 5 years) [91ICR1] with the further provision that the effective dose
should not exceed 50 mSv in any single year. The limit on equivalent dose for the lens of the eye is
150 mSv in a year, the limit on equivalent dose for the skin is 500 mSv in a year. It is implicit in these
recommended dose limits that the dose constraint for optimization should not exceed 20 mSv in a year.
The limits on effective dose for apprentices and students aged between 16 and 18 years who, in the course
of their studies are obliged to use radioactive sources, is 6 mSv per year. Special protection is required
during pregnancy: the equivalent dose to the child to be born is limited to 1 mSv to the reminder of the
duration of pregnancy (see also Sect. 4.8).
The decision to provide individual monitoring for an individual or a group of workers depends on
three major factors: the expected dose in relation to the constraint or limit, the likely variations in the dose
in time and space, and the complexity of the measurement and of the interpretation procedures. Individual
monitoring is required for category A workers. It should be established and monitored by an approved
dosimetric service. Category A includes any radiation work in which the annual effective dose is or might
be higher than 6 mSv, or the annual equivalent dose of the lens of the eye, the skin or hands and feet is or
may be higher than 3/10 of the dose limit stipulated for these tissues or organs. Category B includes all
other radiation work. For practical reasons, monitoring of category B workers is often treated similar to
category A workers by individual monitoring.
In many cases the individual monitoring of external exposure is fairly simple and does not require a
heavy commitment of local resources (see 10.2.6). However, in mixed radiation fields, e.g.
neutron/photon fields, monitoring is much more complex (see 10.2.6). For special groups of workers, e.g.
to the air craft crew, doses caused by cosmic radiation are determined by calculations based on their
flying hours and the flight plans rather than by individual measurement.
In situations where individual monitoring is not appropriate or feasible the occupational exposure
shall be assessed on the basis of the results of area monitoring at the workplace and on information on the
location within the area considered and the duration of exposure.
The control of occupational exposure can be simplified by the designation of work places as
controlled and supervised areas. Controlled Areas are subject to special rules for the purpose of
radiation protection and to which access is controlled. In Supervised Areas, a minimum radiological
surveillance of the working environment will be organized. Outside these designated areas, the dose rates
from sources and the risk of contamination by unsealed radioactive material will be low enough to ensure
that the level of protection for those who work in the premises will be comparable with the level of
protection required for the public.
In several areas of medicine the control of occupational exposures is of particular importance, e.g.
nursing of brachytherapy patients, palpation of patients during fluoroscopy, and interventional radiology
[96ICR]. In these cases individual monitoring with careful scrutiny of the results is always important.
10.2.3.2 Public exposure
The control of public exposure in all normal situations is exercised by the application of control at the
source. Almost all public exposure is controlled by the procedures of constrained optimization and the use
of dose limits. In particular, appropriate monitoring equipment and surveillance programs are required
from the licensee to assess public exposure related to any practice or source and to demonstrate that the
dose to members of the public does not exceed authorized dose limits.
This includes environmental monitoring systems measuring doses or dose rates in the vicinity of a
source or widely distributed in a country for the purpose of surveillance or early warning. Routine
individual monitoring of public exposure is not necessary under normal conditions.
Landolt-Brnstein
New Series VIII/4

Ref. p. 10-39]

10 Measuring techniques

10-31

In all areas of medicine there are no restrictions on the public access to non-designated areas. Public
access to controlled areas should be limited to visitors of patients only, who should be advised of any
restrictions on their behavior.
All reasonable steps shall be undertaken to assess any exposure incurred by members of the public as
a consequence of an accident. This assessment will be based on data of area dosimeters installed in and
around the facility involved, on model calculations based on plant status or on information about any
environmental contamination, and on results of environmental and individual (physical and biological)
monitoring. For practical purposes decisions on interventions are often based on derived secondary limits
of dose rate and on values of the contamination level in agricultural and other environmental products,
which can easily be measured [99IAE].
Emergency response personnel, although not normally occupationally exposed may have to carry out
their duties in areas where there is a potential for elevated radiation exposure. Protection of this personnel
should be treated as part of the occupational exposure incurred in a practice.

10.2.4 Types of monitoring programs


In the context of this Section, two types of monitoring programs will be discussed, e.g. individual and
area monitoring. In many cases where photon radiation is dominant both programs can be considered as
independent means to estimate the effective dose a person would receive or has received. In situations
with significant contribution of -radiation or in mixed neutron-photon radiation fields, data and
information from both types of monitoring programs might be required to arrive at reliable estimates of
the total dose.
10.2.4.1 Individual monitoring for external exposure
External dosimetry deals with radiation that originates outside the body. The external exposure may result
from photon irradiation (X- or -rays), particle irradiation (electrons, neutrons, protons, heavy particles)
or from mixed irradiation (e.g. -rays and neutrons). The exposure may involve the whole body or may be
confined to a sizeable part of the body. It may be localized, from a narrow beam irradiation or a small
radiation source near to the body.
The design of a monitoring program should include the specification of the type(s) of dosimeter to be
used and how and where they should be worn (see Sect. 10.2.5). In complex and inhomogeneous fields it
will often be necessary to use more than one dosimeter. In particular, operations involving manipulations
of radioactive sources may call for dosimeters worn on the fingers. In radiation fields with both
penetrating and weakly-penetrating radiations, e.g. - and -rays, a two component dosimeter is required.
Sometimes neutrons may contribute to the total dose from occupational exposure. In situations where
neutron exposures are likely to significantly contribute to the effective dose special neutron dosimeters
are necessary for monitoring. A detailed overview on individual monitoring of external radiation is given
in [01Bar].
In the case of radiological accidents with low external exposures only, the assessment of effective and
equivalent dose would be covered by routine monitoring programs. In cases where highly elevated dose
levels can not be excluded additional dosimeters, preferentially with direct reading of the dose and dose
rate and with the option of an audible or acoustical warning should be considered.
10.2.4.2 Area monitoring for external exposure
The purpose of area monitoring at workplaces is to ascertain that a working area is free of significant
levels of radiation and contamination. Area monitoring allows the warning of personnel to avoid
hazardous areas. The nature and frequency of workplace monitoring shall be sufficient to evaluate the
Landolt-Brnstein
New Series VIII/4

10-32

10 Measuring techniques

[Ref. p. 10-39

radiological condition at the workplace and to assess the exposure in controlled areas. The routine
workplace monitoring program will usually involve the use of repeated survey measurements. Such a
program may include the use of continuously operating monitors installed at fixed and representative
locations to monitor the normal radiation level and to identify the onset of abnormal or emergency
conditions. The frequency with which routine monitoring will be conducted is determined by the stability
of the radiation environment. If the radiation fields are liable to increase rapidly and unpredictably to
significant levels, the monitoring program should include instrumentation with technical provisions for
fast early warning.
Most of the instruments used at the workplace will measure dose rate rather than dose. Particular care
is required in the selection and calibration of instruments used to measure neutrons, -rays and low
energy photons.
Task related monitoring will provide forecast of the doses likely to be accumulated during a task. For
this purpose portable instruments will preferably be used. Particular care is needed when working with
beta and other weakly penetrating radiations. Special care should be given to the measurement of the dose
rate adjacent to surface or point sources.
10.2.4.3 Calibration
Calibration aims at establishing the relationship between values indicated by a measuring instrument or
system (see Sect. 10.1) and the corresponding true values of a quantity to be measured. The radiation
types used for the calibration of dosimeters are mainly photons, neutrons and beta particles. Calibrations
for each of these types are performed differently using different instrumentation and techniques.
Calibration should closely follow the recommendations of the International Organisation for
Standardisation (ISO) dealing with reference radiations and be based on the methods described in these
standards [96ISO1, 96ISO2, 97ISO, 98ISO, 99ISO, 00ISO1, 01ISO]. A detailed description of the
calibration procedures can be found in [94Alb, 00Die].
The calibration of personal dosimeters or area survey meters used for radiation protection purposes is
mostly a three step process. First, the value of a physical quantity such as air kerma rate or particle
fluence rate of which primary standards usually exist, is determined by a reference instrument at a
reference point in the radiation field used for calibration. Second, the value of the appropriate operational
quantity is determined by application of a conversion coefficient relating the physical quantity to the
radiation protection quantity. Conversion coefficients used to determine operational quantities for
neutrons and photons were evaluated by international committees and finally accepted for general use by
international agreements (see Sect. 6.12, 6.3 and 6.4). Third, the device being calibrated is placed at this
reference point to determine the response of the instrument to the operational quantity, e.g. the personal,
ambient or directional dose equivalent or their corresponding rates. While area dosimeters are generally
calibrated free in air, personal dosimeters are always calibrated in front of a standardized phantom (details
see Sect. 4.5.3.4).
The primary physical quantity used to specify a photon radiation field is exposure or air kerma, and
the primary standard instruments used for its measurement are air-filled ionization chambers. For photon
energies up to about 150 keV, mostly a free-air chamber is used as a standard instrument to measure air
kerma. For higher photon energies, air-equivalent walled cavity chambers are generally employed.
Properties of radiation fields used for the calibration of photon dosimeters are described in ISO standard
4037 [96ISO1].
Calibrations of dosimeters and survey instruments for the measurement of beta radiation are
performed using standard reference beta sources as specified in ISO standard 6980 [96ISO2].
Determination of the conventional true value of the absorbed dose, and hence the directional dose
equivalent, is achieved with a thin-window extrapolation ionization chamber [97Amb].
The primary quantity measured for neutrons is the fluence. In monoenergetic neutron fields the
fluence is measured either directly by a reference instrument (e.g. proton recoil telescope, proportional
counter or Long Counter) or by applying the associated particle method. As regards radioactive neutron
sources, e. g. 252Cf without or with a surrounding D2O-sphere, the neutron fluence is determined from the
source emission rate which is usually determined from comparative activation measurements performed
Landolt-Brnstein
New Series VIII/4

Ref. p. 10-39]

10 Measuring techniques

10-33

by a national standards laboratory. The emission rate is then used to compute the neutron fluence or
fluence rate. In addition, the neutron energy spectrum must be known. With the known spectral fluence
mean conversion coefficients can be calculated and applied to determine the ambient dose equivalent at a
reference point [98ISO] (see Sect. 6.4).
The calibration of a personal dosimeter or area survey meter is not complete without the calibration
being documented. National regulations often specify the details and format of both calibration records
and certificates, as well as the frequency of calibration and the period of time for which calibration
records are to be kept. The following list gives a general guideline for calibration records or certificates.
A certificate should include:
1.
2.
3
4.
5.
6.
7.
8.

Date and place of calibration,


Description of dosimeter or instrument (type and serial number),
Owner of device,
Descriptions of reference radiation sources and standard instruments,
Reference conditions, calibration conditions or standard test conditions,
Results with statement of uncertainties,
Names of the person who performed the calibration and of the reviewer,
Any special observations.

10.2.5 Requirements for individual monitoring of external exposure


The basic requirement for personal dosimeters are to provide a reliable measurement of the
appropriate quantities, i.e. Hp(10) and Hp(0.07) for almost all practical situations, independent of type,
energy and direction of incidence of the radiation and with prescribed overall accuracy. These basic
requirements can be expressed in terms of operational and technical parameters influencing the
performance of the dosimeter, e.g. its response to radiation type, spectral and directional distribution,
environmental influences and practical aspects. The most important ones are described below.
The following minimum requirements apply to all types of personal dosimeters:

convenient in size and shape, low weight, easy to wear


inexpensive
mechanically robust
easy to handle
adaptable to various applications (assessment of whole body dose or extremity dose)
a broad range of doses should be measured
response should be reasonably independent of the radiation energy and dose rate
response should not be strongly influenced by normal changes of environmental conditions
(minimum temperature range 10 C to 40 C, relative humidity: 10 to 90 %)
the measured dose should not be influenced by other unconsidered types of radiation
the dosimeter reading should be independent of any delay between irradiation time and time of
evaluation.

Electronic personal dosimeters are usually capable to measure dose and dose rate and additionally
include an immediate warning capability.
For extremity dosimeters where, due to the close proximity to the source large variations in the dose
rate may occur, small-sized detectors such as detectors kept in finger rings are required (cf. Fig. 10.16).
Detailed technical requirements are specified for detectors suitable for measuring whole body
exposures and partial exposures in [94EU]. Some are summarized in Tables 10.6 and 10.7.

Landolt-Brnstein
New Series VIII/4

10-34

10 Measuring techniques

[Ref. p. 10-39

Table 10.6. Technical requirements for measuring whole body exposures


Photons
Neutrons
Measurement range
Max. range
Measurement range Max. range
Dose range
0.2 mSv to 1 Sv
up to 10 Sv
0.2 mSv to 1 Sv
up to 10 Sv
10 keV
103 keV to 1.5 MeV up to
Energy range(s) 15 keV to 250 keV
or
to
or
100 MeV
70 keV to 1.5 MeV
10 MeV
1.5 MeV to 15 MeV
Dosimeter
0 to 60
0 to 180
0 to 60
0 to 180
orientation
Table 10.7. Technical requirements for measuring partial body exposures
Photons
Beta radiation
Measurement range
Max. range
Measurement range
Max. range
Dose range
1 mSv to 10 Sv
0.1 mSv to 10 Sv 1 mSv to 10 Sv
0.1 mSv to 10 Sv
15 keV to 1.5 MeV
10 keV to 10 MeV 0.2 MeV to 0.5 MeV1) 0.06 MeV to
Energy range
1.0 MeV1)
Dosimeter
0 to 60
0 to 180
0 to 60
0 to 180
orientation
1)

mean beta energy

10.2.5.1 Operational requirements


Two principal questions have to be answered before a measurement of Hp(10) and Hp(0.07) can be
interpreted for radiation protection purposes. The personal dosimeter has to be worn at a representative
place on the surface of the body and it will measure the dose at this point for a predefined period of time.
In many practical cases the radiation field will be inhomogeneous and multidirectional. The value
measured will therefore often depend on the orientation of the body in the field. While the use of several
dosimeters could in principle improve the situation and lead to a more representative assessment of the
effective dose, mostly one dosimeter measuring Hp(10) is sufficient for routine monitoring.
In order to measure Hp(10) for assessment of effective dose, a personal dosimeter is usually worn in
front of the body between the shoulders and the waist. Dosimetry of the skin dose or an extremity dose
can be performed by using finger batches worn on the hand as rings. An estimate of the eye equivalent
dose is obtained by wearing a whole body dosimeter placed at the collar. This is a reasonable location to
measure both the eye and the whole body dose. Alternative locations for dosimetry may be necessary in
the course of certain types of work. An example is the use of lead aprons in X-ray applications. In this
case two dosimeters may be necessary, one under the apron to measure Hp(10) and one at the collar to
measure the dose to the head. Other situations may necessitate relocation of dosimeters including fetal
monitoring where the monitor should be placed in front of the abdomen to assess the uterus or fetal dose.
In many practical cases of routine monitoring, dosimeters will be worn over a period of one month. In
low dose environments this period could be extended up to 6 months. In cases with highly variable
radiation fields and in situations where there are indications that dose limits could be reached or
exceeded, an evaluation of the dose within a shorter time period could be required. If for operational
reasons daily monitoring is required, a direct reading dosimeter with sufficient sensitivity should be used
in addition to the routine dosimeter. Direct reading dosimeters are frequently used to monitor the dose
received during a particular task, e.g. one working day or one shift. In the case of a pregnant woman an
electronic dosimeter would be an appropriate way of individual monitoring (cf. Fig. 10.13).

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-39]

10 Measuring techniques

10-35

10.2.5.2 Accuracy requirement


Basic rules in the description of uncertainties in measurements are given in a joint document of BIPM/
/IEC/ISO/OIML [95ISO]. The errors and uncertainties in the use of monitoring to provide estimates of
individual doses and intakes result from the measurements and from the models used to link the measured
and the required quantities. Different types which contribute to the overall uncertainties can be
distinguished: random uncertainties due to counting statistics, systematic errors due to calibration errors
and errors in dosimetric and metabolic models and errors in practical application of the models. For most
assessments, the systematic errors in modelling result in a bias towards over-estimation of the true dose.
Basic recommendations on the acceptable uncertainty in routine individual monitoring are given by
ICRP Publication 60 (par. 271) [91ICR1], Publication 75 [97ICR] and by ISO [00ISO2]: for annual doses
of the order of the relevant annual limit, the apparent annual dose to an individual as indicated by routine
dosimeters should not differ from the annual dose equivalent indicated by an ideal dosimeter by more
than 33 % or +50 % at the 95 % confidence level. The 95 % confidence level means that the given
requirement must be fulfilled for 19 of 20 different measurements. For dose values equal to or close to the
annual dose limit, e. g. 20 mSv (see 10.2.3.1), the relation between the measured and the true value may
thus vary between 1.5 and 1/1.5 times this value in 19 of 20 different measurements.
For individual doses much below the annual limit the accuracy requirements are less than the values
given above. For characterising the acceptable uncertainty in performance tests of individual dosimeters
trumpet curves as proposed by Bhm et al. [90Boe] have been defined by ISO [00ISO2] describing the
requirement by an interval for Hm/Ht (measured dose value/conventionally true dose value) as a function
of dose. A detailed overview on requirements for photon dosimeters and dosimetry services as published
in the various international recommendations and standards is given by Ambrosi et al. [98Amb, 01Amb].
As example, Fig. 10.26 shows the recommendation of the European Commission [94EU] and the IAEA
[97IAE] for photon dosimeters. While many countries, e.g. Germany, Italy, Sweden and Switzerland, use
this procedure in performance tests, other countries, e.g. Spain, UK and USA, uses a statistical evaluation
of the measured values together with criteria for a bias setting [01Amb].
2.0

Measured dose / true dose Hm / H t

Measured dose / true dose Hm / H t

2.0

1.5

1.0

0.5

10 -2

1
10 -1
10
Dose equivalent Hp (10) [mSv]

1.5

1.0

0.5

0
10 -2

10 2

10 -1
1
10
Dose equivalent Hp (10) [mSv]

10 2

Fig. 10.26. Requirements in performance tests of individual dosimeters for photons showing upper and lower
limits for the ratio of the measured dose to the true dose value, Hm/Ht, as a function of dose [90Boe, 98Amb]
where 95 % of all measured values must be within these limits. Full lines: monthly monitoring period; broken
lines: bi-weekly monitoring periods. (a) limits for Hp(10); (b) limits for Hp(0.07).

The accuracy requirements for individual dosimeters for neutrons are mostly less than those for
photon dosimeters because of the difficulties in realising a dosimeter response sufficient independent of
neutron energy (cf. Sect. 10.1.7). Often it may be necessary to use some information about the spectral
Landolt-Brnstein
New Series VIII/4

10-36

10 Measuring techniques

[Ref. p. 10-39

distribution of the neutrons in order to apply an approximate calibration factor to the dosimeters used. In
situations where the contribution of the neutron dose is substantial and where the total dose may approach
the dose limit, the radiation field should be characterised by application of more sophisticated equipment,
e.g. neutron spectrometry, to get better information about the neutron spectrum and its directional dependence.

10.2.6 Personal dosimeters for individual monitoring in different radiation fields


The most abundant types of detectors used for the purpose of personal dosimetry in routine monitoring
are the film dosimeter, the photoluminescence (glass) dosimeter (PLD), thermoluminescence dosimeters
(TLD) and ionization chambers. Also electronic dosimeters, based e.g. on GM-counters, proportional
counters or semiconductor detectors (see Sect. 10.1), are used. Film dosimeters and ionization chambers
are used to monitor X-, - and high energy -ray exposures by measuring Hp(10). Glass and TLDs are
able to measure both Hp(10) and Hp(0.07). They are used to monitor -, -, X-rays, and neutron radiations.
Etched-track detectors and bubble detectors measuring Hp(10) are mainly sensitive to neutrons only.
Small TLD badges, e.g. finger badges are designed to be worn on the finger to record the dose to the
hand. They are sensitive to X- and -rays and high energy beta rays. A detailed description of the various
detector types is given in Sect. 10.1.
In many practical situations where task monitoring is required in environments with the possibility of
elevated radiation levels, active dosimeters are needed which include immediate warning capabilities,
e.g. electronic dosimeters. Advanced methods of active neutron dosimetry in mixed radiation fields are of
particular importance. Potential fields of application of active dosimeters of this kind are at nuclear power
plants and particle accelerators.
10.2.6.1 Photon dosimetry
In most work situations with exposures by strongly penetrating electron/photon radiation, an estimate of
Hp(10) can be obtained from a single dosimeter sensitive to electrons and photons. The overwhelming
share of occupational exposure is caused by photons. Mostly film or TL dosimeters are used in routine
monitoring, sometimes also PLDs are in use [01Bar]. Since 1980, the application of TLDs has increased
considerably. For partial body dosimeters nearly always TLDs are used. In a few cases where the
workers doses are at or near the limits, it may be necessary to obtain additional information about the
exposure conditions, e.g. from field measurements at the workplace to better estimate the effective dose
equivalent.
Film dosimeters (Sect. 10.1.6) as compared with solid state dosimeters involve a somewhat greater
uncertainty of measurement in the lower dose range (<0.4 mSv) in the case of very hard gamma radiation.
Test measurements have shown that all three dosimeter types fulfill the performance requirements
mentioned in Sect. 10.2.5.2 (see Fig. 10.27). If well designed the dosimeter types are suitable for dose
measurements up to photon energies of 15 MeV. Their response to neutrons with energies up to about
1 MeV is usually very low, i.e. they measure the photon dose independent of the neutron dose.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-39]

10 Measuring techniques

10-37

2.0

HMST / H PTB

1.5

1.0

0.5

10 -1

1
10
10 2
Photon dose H PTB [mSv ]

103

Fig. 10.27. Results of test measurements with different


personal dosimeters. The ratio of the measured dose to the
true dose as a function of photon dose is shown [01Amb].
The lines mark the accuracy limits for a monthly
monitoring period.
Film dosimeter
Thermoluminescence dosimeter
Photoluminescence dosimeter.

10.2.6.2 Beta dosimetry


In most cases the dose generated by weakly penetrating beta radiation is a partial body dose of the skin of
uncovered extremities. Although the depth of the sensitive layer of the skin vary between individuals and
over the body of individuals, Hp(0.07) is considered to be a reasonable quantity to apply for the
assessment of doses if the dosimeter is worn at a position representative for the exposure. Monitoring of
weakly penetrating radiation is predominantly achieved by partial body dosimeters with thermoluminescence probes. In the case of hard beta radiation, e.g. from 90Sr/90Y or 204Tl, the dose is detected
with a sufficient degree of reliability with film dosimeters. Sensitivity and energy-dependence of the film
are sufficient for all practical purposes even in the low dose range. For a detection of beta radiation with
intermediate energies above 100 keV the film dosimeter is, in principle, well suited. In practice, most
radiation fields are mixed photon/beta fields and when measuring Hp(0.07) correctly it is generally
difficult to obtain a separate assessment of the fractions of the dose from photon and beta radiation. In this
case an interpretation of the measured data must be based on additional information by the licensee.
The detection of weakly penetrating beta radiation to the skin of the hands with standard finger
dosimeters is not always satisfactory. Many of these dosimeters are intended to be used for measuring
photon radiation. They may considerably underestimate exposures from beta radiation. Sensitive thin
layer thermoluminescence dosimeters need to be used for this purpose. In most practical situations the
skin will be exposed to weakly penetrating radiation together with strongly penetrating radiation and an
estimate of the skin dose will have to take account of both types of radiation.
10.2.6.3 Neutron dosimetry
Personal dosimeters for neutrons have not yet reached the quality standards of photon dosimetry. This is
mainly because their sensitivity and their variation of response with neutron energy and with the angle of
radiation incidence are unsatisfactory. The nuclear track film which has frequently been used is only
suited for fast neutrons with energies >1 MeV. A special type of thermoluminescence probes, e.g. the
albedo dosimeter, is suitable to measure both the photon and the neutron dose. The photon energy range
of this dosimeter type is 15 keV to 10 MeV, for neutrons from thermal neutrons up to 20 MeV while the
response to neutrons, however, is strongly decreasing with neutron energy (see Sect. 10.1.7).
A reasonable approach for individual neutron dosimetry is to use more than one type of detector to
cover the whole energy spectrum, e.g. an Albedo dosimeter for neutrons in the low energy region together
with a solid state etched-track dosimeter to cover the energy range above approximately 100 keV
Landolt-Brnstein
New Series VIII/4

10-38

10 Measuring techniques

[Ref. p. 10-39

(cf. Sect. 10.1.7). Even with this detector system neutrons with intermediate energies may not be
measured to full satisfaction. More recently, electronic personal dosimeters for neutrons based on
semiconductor devices became available which partially improve the situation. In cases where the neutron
dose contributes significantly to the total dose and the total dose is likely to approach dose limits, a more
elaborate approach may be necessary. In such situations the use of area monitors and of neutron
spectrometers is recommended to better characterize the radiation field.
10.2.6.4 Dosimetry in mixed field situations (photons and neutrons)
In a mixed field situation with photons and neutrons the personal dose equivalent, Hp(10), includes the
contributions of both photon and neutron dose. In mixed field situations an improvement of the measuring
methods is still required. There are several ways to estimate the total dose and some commercial devices
exist to perform appropriate measurements. Either two dosimeters are used each of them sensitive to
photons or neutrons only, or one detector which measures the total dose directly. But often the energy
response of these devices fails where their physical properties would be attractive, i.e. inside the
containment of nuclear power plants, where low energy neutrons dominate. Bubble detectors for neutrons
are available on the market but in the attempt to make them robust their unique response properties have
been neglected. Tissue equivalent proportional counters (TEPC), which measure the total dose equivalent
in a mixed field rather than being just neutron dosimeters have been developed but without real
breakthrough on the market.
There have been ongoing developments in recent years [99Alb] and a few most promising monitoring
techniques may be ready for routine application in near future. The TEPC personal dosimeter can
determine neutron dose equivalents down to 10 Sv with sufficient accuracy. The dosimeter offers the
option of a detailed quantification of any radiation exposure in terms of a microdosimetric spectrum.
Combined neutron and photon dosimeters of very small dimension, e.g. ionization chambers with direct
ion storage (DIS) are recommended for application in radiation fields with high contributions of neutron
doses and in places where light-weight and small dimensions are important because of the type of work
performed. Dosimeters based on superheated drop detectors reveal a high sensitivity to neutrons with no
sensitivity to photons.

Landolt-Brnstein
New Series VIII/4

10 Measuring techniques

10-39

10.2.7 References for 10.2


77ICR
85ICR
90Boe
91ICR1
92ICR
93ICR
94Alb

94EU
95ISO
96EU
96ICR
96ISO1

96ISO2
97Amb
97IAE
97ICR
97ISO

98Amb
98ICR
98ISO

International Commission on Radiological Protection: ICRP Publication 26. Oxford, UK:


Pergamon Press, 1977.
International Commission on Radiation Units and Measurements: ICRU Report 39, Bethesda,
MD: ICRU Publications, 1985.
Bhm, J., Ambrosi, P.: Mandatory type tests of solid state dosimetry systems as an appropriate
aid to quality assurance in individual monitoring. Radiat. Prot. Dosim. 34 (1990) 123-126.
International Commission on Radiological Protection: ICRP Publication 60. Oxford, UK:
Pergamon Press, 1991.
International Commission on Radiation Units and Measurements: ICRU Report 47. Bethesda,
MD: ICRU Publications, 1992.
International Commission on Radiation Units and Measurements: ICRU Report 51. Bethesda,
MD: ICRU Publications, 1993.
Alberts, W.G., Bhm, J., Kramer, H.M., Iles, W.J., McDonald, J., Schwartz, R.B., Thompson,
I.M.G: International standardisation of reference radiations and calibration procedures for
radiation protection instruments. Proc. German-Swiss Radiation Protection Association
Meeting 1994, Karlsruhe, 1994.
European Commission: Radiation Protection 73, Technical recommendations for monitoring
individuals occupationally exposed to external radiation. EUR 14852 EN, EC, Luxembourg,
1994.
International Organization for Standardization: Guide to the expression of uncertainty in
measurement. Geneva, Switzerland: ISO, 1995 (corrected of first print in 1993).
Council Directive 96/29/EURATOM of 13 May 1996 laying down basic safety standards for
the protection of the health of workers and the general public against the dangers arising from
ionising radiation; EC Journal Series L 159 (1996)
International Commission on Radiological Protection: ICRP Publication 73. Oxford, UK:
Pergamon Press, 1996.
International Organization for Standardization: X and gamma reference radiations for
calibrating dosemeters and dose rate meters and for determining their response as a function of
photon energy. Part 1: Radiation characteristics and production methods. ISO/4037-1, Geneva,
Switzerland, 1996.
International Organization for Standardization: Reference beta-radiations for calibrating
dosimeters and dose rate meters and for determining their response as a function of betaradiation energy. ISO/6980, Geneva, Switzerland, 1996.
Ambrosi, P.: Improved beta secondary standard; PTB-News 97.1 (1997) 3
IAEA Safety Series: Draft safety guide: Assessment of occupational exposure to external
radiation. NENS-12, IAEA, Vienna, 1997.
International Commission on Radiological Protection: General principles for radiation
protection of workers. ICRP Publication 75. Oxford, UK: Pergamon Press, 1997.
International Organization for Standardization: X and gamma reference radiations for
calibrating dosemeters and dose rate meters and for determining their response as a function of
photon energy. Part 2: Dosimetry for radiation protection over the energy range 8 keV to 1.3
MeV and 4 MeV to 9 MeV. ISO/4037-2, Geneva, Switzerland, 1997.
Ambrosi, P., Bartlett, D.: Dosimeter characteristics/ Dosimeter and service performance
requirements. PTB-Dos-27, PTB, Braunschweig, 1998.
International Commission on Radiation Units and Measurements: Conversion coefficients for
use in radiological protection against external radiation. ICRU Report 57, Bethesda, MD:
ICRU Publications, 1998.
International Organization for Standardization: Reference neutron radiations - Part 3:
Calibration of area and personal dosimeters and the determination of their response as a
function of neutron energy and angle of incidence. ISO/8529-3 Geneva, Switzerland, 1998.

Landolt-Brnstein
New Series VIII/4

10-40
99Alb

99IAE
99ISO

99Zan
00Die
00ISO1
00ISO2
01Amb
01Bar
01ISO

10 Measuring techniques
Alberts, W.G., Arend, E., Barelaud, B., Curzio, G., Decossas, J.L., dErrico, F., Fiechtner, A.,
Grillmaier, R., Meulders, J.-P., Menard, S., Roos, H., Schuhmacher, H., Thevenin, J.-C.,
Wernli, C., Wimmer, S.: Advanced methods of active neutron dosimetry for individual
monitoring and radiation field analysis (ANDO), Report PTB-N-39, Braunschweig, 1999.
Generic procedures for monitoring in a nuclear or radiological emergency, IAEA-TECDOS1092, Vienna, 1999.
International Organization for Standardization: X and gamma reference radiations for
calibrating dosemeters and dose rate meters and for determining their response as a function of
photon energy. Part 3: Calibration of area and personal dosemeters and the measurement of
their response as a function of energy and angle of incidence. ISO/4037-3 Geneva,
Switzerland, (1999).
Zankl, M.: Personal dose equivalent for photons and its variation with dosimeter position.
Health Phys. 76 (1999) 162.
Dietze, G.: Dosimetric concepts and calibration of instruments, IRPA 2000, Hiroshima, May
2000.
International Organization for Standardization: Reference neutron radiations - Part 2:
Calibration. Fundamentals of radiation protection devices related to the basic quantities
characterizing the radiation field. ISO/8529-2 Geneva, Switzerland, 2000.
International Organization for Standardization: Radiation protection criteria and performance
limits for the periodic evaluation of personal dosemeters for x and gamma radiation.
ISO/14146, Geneva, Switzerland, 2000.
Ambrosi, P.: Dosimetric performance requirements for the routine dose assessment of external
radiation. Radiat. Prot. Dosim. 96 (1-3) (2001) 67-72.
Bartlett, D.T., Bhm, J., Hyvnen, H. (eds): Individual monitoring of external exposure. Proc.
Europ. Workshop 2000. Radiat. Prot. Dosim. 96 (1-3) (2001).
International Organization for Standardization: Reference neutron radiations - Part 1:
Characteristics and Methods of Production. ISO/8529-1 Geneva, Switzerland, 2001.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-51]

10 Measuring techniques

10-41

10.3 Radiological protection measurements: Internal exposure


10.3.1 Measurement of radon and its progeny
Radon (222Rn) and thoron (220Rn) are gaseous radionuclides in the U- and Th-decay chain, respectively,
occurring naturally in the ground and escaping from there into air and water (see Chapter 11). Their decay
products are metallic radionuclides (see Tables 10.8 and 10.9). In air usually a mixture of radon/thoron
and short-lived radon/thoron progenies exist. The progenies are mostly attached to aerosols with sizes of
about 0.01 - 10 m in diameter. Some (few percent only), however, are non-attached (cluster
<0.005 m in diameter). Due to the short half-life of 55 s of thoron this nuclide and its decay products are
less important for dose from inhalation than radon (T1/2 = 3.825 d) and its short-lived progeny. In special
situations, however, they may contribute to the total potential alpha energy concentration from radon and
thoron progenies up to 50 %.
While the inhaled radon is mostly exhaled again, the progeny are deposited in the respiratory tract
where their decay by alpha-particle emission is seen to be most relevant for the dose to the lung and hence
for lung cancer induction. Their contribution to the dose is generally 2 to 3 orders of magnitude greater
than that of 222Rn. In measurements, however, the radon is given the highest interest, because action
levels and reference values are nearly always specified in terms of radon concentration (in Bq m3) or
radon exposure (in Bq m3 h) and not in terms of dose quantities (see Sect. 4.8). If doses wanted to be
specified, the radon progeny concentrations in air or the equilibrium factor F need to be determined. For
radon in homes often an equilibrium factor F = 0.4 is applied, if no measurements are available.
Table 10.8. 226Ra decay chain with radon and its progeny (data from [98NN]).
Radionuclide
Half-life T1/2 Radiation energy and relative emission probabilities
-particles
-particles(1)
-rays
MeV
(%)
MeV
(%)
MeV
(%)
226
4.59
(4.16)
0.186
(3.51)
Ra
1 600 a
4.78
(94.5)
222
Rn
3.825 d
5.49
(100)
218
Po
3.10 min
6.00
(100)
214
Pb
26.8 min
0.67
(48)
0.242
(7.4)
0.73
(42)
0.295
(19.3)
1.02
(6)
0.359
(37.6)
other
(4)
214
Bi
19.9 min
1.00
(23)
0.609
(46.1)
1.51
(40)
1.120
(15.1)
3.26
(19)
1.764
(15.4)
other
(18)
214
Po
7.69
(100)
164 s
210
Pb
22.3 a
0.015
(81)
0.047
(4.05)
0.061
(19)
210
Bi
5.013 d
1.161
(100)
210
Po
138.4 d
5.30
(100)
206
Pb
stable
(1)

The energy given is the maximum energy of -particles emitted in the specific decay channel.

Specific quantities have been defined taking care of the complex decay chain of radionuclides (see
Sect. 3.4.1) and the importance of the progeny for internal dosimetry. While in measurements of
radon/thoron the actual activity concentration or its mean value over a longer period is usually
determined, for the progeny the potential alpha energy concentration (PAEC), the equilibrium equivalent
concentration (EEC), the equilibrium factor and the potential alpha energy exposure are measured (see
Sect. 4.6).

Landolt-Brnstein
New Series VIII/4

10-42

10 Measuring techniques

[Ref. p. 10-51

For radon the concentration in air above ground, in the air, in the ground, and in water from the
ground are of interest, while the progeny are important only when produced in air above ground. In
buildings often the exhalation rate of radon from the ground or wall materials is looked at. This can be
determined by measuring the increase of the radon concentration in a closed volume after it has been well
ventilated.
The diffusion of radon in air is very fast and its concentration in a closed room is, therefore, usually
homogeneous. Mostly there is no equilibrium with its progeny because of a continuous deposition of
aerosols on the walls and other surfaces. Often an equilibrium factor between 0.3 and 0.7 is achieved.
Local radon concentrations in air, however, may strongly vary with time depending on environmental
parameters like weather conditions, pressure, wind, temperature as well as ventilation in rooms. Care
must, therefore, be taken when using data from short time measurements. Also the positioning of
instruments for measurement of radon needs attention in order to avoid erroneous results. In rooms
detectors should usually not be positioned near windows or doors or directly on a wall.
For thoron, the situation is generally different. Free in air, there is mostly a strong decrease of the
thoron concentration with height above ground due to the short half-life of 55 s, and equilibrium with
thoron progeny is never achieved.
Table 10.9.

228

Th decay chain with thoron and its progeny (data from [98NN]).

Radionuclide

Half-live T1/2

228Th

1.913 a

224

3.66 d

Ra

220

Rn
Po
212
Pb

55.6 s
0.15 s
10.64 h

212

Bi

60.6 min

212

Po
Tl

304 ns
3.04 min

Pb

stable

216

208

208

Radiation energy and relative emission probabilities


-radiation
-radiation(1)
-radiation
MeV
(%)
MeV
(%)
MeV
(%)
0.0837 (1.2)
5.34
(27.0)
0.216
(0.25)
5.43
(73.0)
5.45
(4.9)
0.241
(4.1)
5.68
(95.1)
6.29
(100)
0.55
(0.1)
6.78
(100)
0.331
(83)
0.239
(43.3)
0.569
(12)
0.300
(3.3)
other
(5)
6.05
(25)
1.55
(5)
0.040
(1.1)
6.09
(10)
2.26
(55)
0.727
(6.6)
other
(40)
1.620
(1.5)
8.78
(100)
1.28
(23)
0.511
(22.6)
1.52
(22)
0.583
(84.5)
1.80
(51)
0.860
(12.4)
other
(4)
2.614
(99.1)

(1) The energy given is the maximum energy of -particles emitted in the specific decay channel.

Radon/thoron and its short-lived progeny form a decay chain and the relation of their activity
concentrations is generally described by a set of differential equations (see Sect. 3.4.1.4). If, for example,
a closed chamber is filled only with radon with an activity A at a time t = 0, the activity of the progeny
will increase from 0 Bq until after about 3 h it reaches an equilibrium (F = 1), where the activities of the
progeny equal that of radon (see Fig. 10.28).
Another example may be the build up and decrease of the activity of radon progeny absorbed in a
filter flowed by air during a fixed period. In this case, the activities of the decay products on the filter are
described by a set of differential equations:
dAi dt = ciV& + i ( Ai-1 Ai )

i=1-4

(10.3.1.1)

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-51]

10 Measuring techniques

10-43

with Ai activity of radionuclide i on the filter in Bq, ci activity concentration of radionuclide i in air in
Bq m3, V& air flow through the filter in m3 h1 and i decay constant of radionuclide i in h1. Fig. 10.31
shows a typical example. The same equations can be used when the activity A is exchanged by the
number of the corresponding particles N.
1200
1000

Activity [Bq ]

800
3

600
400
200

30

60

120
90
Time [min ]

150

180

Fig. 10.28. Example for the decay of radon and built up


and decay of radon progeny in a closed chamber.
Activity at t = 0: ARn-222= 1000 Bq, APo-218 = APb-214 =
ABi-214 = APo-214 = 0 Bq.
(1) 222Rn, (2) 218Po, (3) 214Pb , (4) 214Bi and 214Po.

Different types of measurements are performed in the assessment of radon and radon progeny
concentrations. Measurements within a period of up to 2 days are called short-time measurements. They
are performed by either a sequence of single short measurements, by a continuous measurement or by a
measurement with an integrating device. Such measurements provide the actual radon concentration at a
place of interest but are usually not sufficient for the estimation of a representative mean annual or
monthly value because of the possible environmental variations.
Long-term measurements ranging for some months or a year are usually performed with integrating
passive detectors which at the end deliver a value integrated over the selected period and hence a mean
radon concentration which can be related to reference values, action levels or annual limits.
Continuous measurement means that an instrument is continuously measuring and periodically
delivering data where the smallest time period depends on the instrument and the accuracy needed. Those
measurements are performed with active electronic devices and are used if short-time or daily variations
are investigated.
While radon and thoron are mainly emitting -particles, their various progenies decay by emission of
- or -particles accompanied by -rays (see Tables 10.8 and 10.9). Therefore, depending on the aim and
type of measurement very different detector systems are applied, ranging from simple and cheap passive
ones e.g. for screening measurements in houses up to detectors with complex electronic devices used as
reference instruments.
In the following an overview is given on the measurement techniques and devices in use for detection
of radon and its progeny (see also [88NC, 88Naz, 02SSK]). For thoron similar instrumentation may be
applied if care is taken of the short half-life of thoron which limits its diffusion time and, for example, has
the effect that a strong decrease of the thoron concentration exists with increasing height above ground.
In any way, care must be taken that devices may be sensitive to both radon and thoron and their
progenies, and then the influence of thoron on measurements of radon should be estimated. Often a
sufficiently long diffusion time through a filter can avoid such problems.

Landolt-Brnstein
New Series VIII/4

10-44

10 Measuring techniques

[Ref. p. 10-51

10.3.1.1 Measurement of radon in air


General
Radon measurements in air are performed in homes or dwellings or at work places as well as outside in
areas where the exhalation from the ground may be high. Most methods for the measurement of the radon
concentration in air are based on the principle that radon is entering a closed chamber or is adsorbed in
carbon material, while the radon/thoron decay products in air which would strongly influence the
measurement are absorbed before entering the system by a filter. This is realized either by pumping air
through the filter to the chamber or by diffusion of radon through the filter. The radiation from radon and
its progeny from the decay of radon in the chamber or in the carbon absorber are then contributing to a
detector signal. If the diffusion time is longer than some minutes, the contribution from thoron to the
detector response is small due to its short lifetime (T1/2= 55 s). Depending on the detector device either the
charge produced in the chamber by ionisation is measured or -particles from both radon and its progeny
or -particles and -rays from its progeny are detected individually.
Passive integrating systems
Passive integrating systems are those which do not allow a continuous or quasi continuous read out but
deliver a value of the radon concentration integrated over a longer time period (in Bq m3 h). These are
either systems with a passive detector, e.g. a detector chamber with an etched track detector (see Sect.
10.1.7) which detects the -particles emitted by the radionuclides in the chamber gas, or an electret
detector, where the discharge of a charged electret by ionisations in the chamber gas is determined by a
voltage measurement [88Kot], or systems with a radon adsorbing material (an activated carbon absorber)
where radon is trapped on the active sites of the carbon beds, and afterwards the -radiation which is
emitted from the radon decay products 214Pb and 214Bi produced in the absorber is measured by a gamma
spectrometer (see Sect. 10.1.4) [84Geo, 90Geo]. All systems are equipped with a filter in front of the
chamber for retaining radon progeny from entering the chamber (see e.g. Fig. 10.29). Because water
vapor and also temperature influences the radon collection efficiency the activated carbon collector
systems should be used indoors only. Table 10.10 gives an overview on different systems with passive
detectors in use and provides some further information. More details about special systems available on
the market are given in a review by George [96Geo] and a report on a European intercomparison of
passive radon detectors [00How]. Depending on the diffusion time of radon through the entrance filter or
plastic foil the system may also be sensitive to thoron or not.
Integrating detectors are usually mailable and relatively cheap. They are often used for medium and
long time measurements in dwellings where knowledge about radon concentration values averaged over 3
to 12 months are most important. The analysis of the detector response is mostly performed at a central
laboratory and needs more expensive additional equipment.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-51]

10 Measuring techniques

Cover

10-45

Cover
Gasket
Filter

Drying agent

Detector

Filter
Carbon absorber

Fig. 10.29. Schematic diagram of passive devices for radon measurement in air; (a) chamber with activated carbon
absorber; (b) diffusion chamber with either an etched track detector or an electret detector.

In principle, also active systems which allow to nearly continuously providing data may be used as an
integrating device. For long term measurements, however, they become much more expensive.
Table 10.10. Integrating systems with passive detectors for the measurement of radon in air
Method

Measured
radiation

Absorption of radon in -rays


a carbon filter
Absorption of radon in -rays or
a carbon collector
-particles
with entrance filter
Diffusion chamber
with entrance filter

-particles

Diffusion chamber
with entrance filter

charge
from
ionisations

Detector

Lower detection
limit

-spectrometer

5 Bq m3
in 0.5m3 air
10 Bq m3
(3 h measurem.)

-spectrometer
or liquid
scintillation
counter
etched track
105 Bq h m3
detector (CR-39,
LR-115 etc.)
electret detector 103 Bq h m3

Influenced
by
air humidity
temperature
air humidity
temperature

Main
application
short time
measurement
short time
measurement
up to 3 d

air pressure,
temperature,

long time
measurement
1-12 months
long time
measurement
1w-1a

air pressure,
dose rate, radiation

Active detector systems


These systems allow a single measurement within a short time period or quasi continuous measurements
to register temporal variations of the radon concentration. All systems are supplied with an entrance filter
for absorption of the progeny from outer air. Usually, the -particles emitted in the chamber (mostly from
progeny produced in the chamber and deposited on the inner chamber wall) are directly measured (see
Fig. 10.30). The use of a scintillation cell, where the walls are coated with a scintillating material (mostly
silver activated ZnS(Ag) powder), is one of the eldest methods (Lucas chamber [57Luc]). Others are
using silicon surface barrier detectors or diffused junction detectors for the detection of -particles. In
other systems the charge or charge pulses from ionisations in the chamber gas are measured [92Bal].
Table 10.11 gives an overview on different systems with active detectors in use and some additional
information. More specific details on devices already used in practice are given by George [96Geo].

Landolt-Brnstein
New Series VIII/4

10-46

10 Measuring techniques

[Ref. p. 10-51

Table 10.11. Active detector systems for the measurement of radon in air
Lower detection
limit

Method

Measured
radiation

Detector

Ionisation chamber
with entrance filter

- and particles

Multi-wire
ionisation chamber
with entrance filter
Scintillation
chamber with
entrance filter
Chamber with
entrance filter,
electrostatic
deposition
Diffusion chamber
with -spectrometry
Two filter method
(also for thoron)

-particles

5 Bq m3
(103 cm3 volume,
3 h measurem.)
5 Bq m3
(103 cm3 volume,
3 h measurem.)
20 Bq m3
(250 cm3 volume,
3 h measurem.)
silicon surface 5 Bq m3
barrier detector (103 cm3 volume,
3 h measurem.)
charge or
charge pulse
measurement
charge pulse
measurement,
-spectrometry
ZnS(Ag)
scintillator

-particles
-particles

activity of
2nd filter

Pump

Main
application

air humidity

single or
continuous
measurement
single or
continuous
measurement
single or
continuous
measurement
single or
continuous
measurement

air humidity
vibration

air humidity

air humidity
silicon surface 100 Bq m3
barrier detector (102 cm3 volume,
3 h measurem.)
activity
10 Bq m3
determination (105 cm3 volume,
of filter, vari10 h measurem.)
ous detectors

-particles

Filter

Influenced
by

single or
continuous
measurement
single
measurement

Drying filter
Pump

Filter

Detector

Chamber wall

ZnS
Quartz glass
PM
Amplifier

Amplifier

HV
Multichannel analyser

Counter

Electrometer

Fig. 10.30. Schematic diagram of active devices for radon measurement in air; (a) Lucas chamber [57Luc] (PM:
photomultiplier); (b) ionisation chamber with charge measurement (HV: high voltage); (c) diffusion chamber with
semiconductor detector.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-51]

10 Measuring techniques

10-47

10.3.1.2 Measurement of radon progeny in air


Nearly all devices for the measurement of radon progeny are using the filter method which can in general
be described by the following. A pump is sucking air through a filter where the aerosols with the radon
progeny are absorbed. A flow meter is measuring the air volume flowing through the filter and the
activity of the filter is determined by measuring the -, - or -radiation emitted from the progeny either
quasi-continuously or after the collection period at different time intervals. As shown in Fig. 10.31 the
activity of 218Po, 214Pb and 214Bi on the filter vary differently with time, while the activity of 214Po equals
that of 214Bi because of the short lifetime (T1/2 = 164 s). From these data the potential alpha energy
concentration (PAEC in J m3 or WL) or the activity concentration of the different decay products can be
calculated using the set of differential equations (see equ. (10.3.1.1)).
14000
12000

Activity [Bq]

10000
8000
6000

Fig. 10.31. Build up and decrease of the activity of


radon progeny deposited on a filter with 100 %
collection efficiency which for 2000 s is flowed by 103
cm3 min1 of air with a radon activity concentration of
1 kBq m3 and an equilibrium factor F = 1.
(A) 218Po (-emission), (B) 214Pb (, -emission), (C)
214
Bi (, -emission) and 214Po (-emission).

4000
A

2000
0

4000
6000
Time [s ]

2000

8000

10000

The sampling unit and the activity measurement unit are either separated or combined in one system
(see Fig. 10.32). The absorbing filter should have a high efficiency. For - or -radiation measurements a
total absorption efficiency of >98 % is sufficient. For -particle detection, however, it is additionally
important that the progeny are adsorbed at the filter surface. This affords membrane filters with pore sizes
of less than 3 m.
Detector

Filter

Detector

FM

Landolt-Brnstein
New Series VIII/4

Electronics

FM

Computer

Electronics

Computer

Fig. 10.32. Schematic diagram of devices for radon


progeny measurements [96Por]
(FM: flow meter, P: pump); (a)
with separated sampling and
detector systems; (b) with a
combined sampling and detector system.

10-48

10 Measuring techniques

[Ref. p. 10-51

The sensitivity of a system depends strongly on the power of the pump. Mostly pumps with flow rates
from 0.1 m3 h1 to 1 m3 h1 are used, but for special short time measurements also pumps with flow rates
up to 100 m3 h1 are applied. Care must be taken that the aerosols are not already be deposited in the
entrance region of the device in front of the filter.
If a flow meter is included in the system, its position is important. On the one hand it should not be
influenced by the pump (e.g. by vibrations), but on the other hand it should not influence the flow in front
of the filter and the deposition of aerosols. Immediately behind the filter might be the best position.
Determination of the activity concentration of the radon progeny
Three independently measured total count numbers for decay by -particles during different counting
intervals after the sampling period can be used to calculate the progeny concentrations using equ.
(10.3.1.1) [96Por]. This method has been developed by Tsivoglou [53Tsi] and was further improved by
various groups [72Tho, 80Bug, 81Sco, 84Naz] in order to improve its application also for low count rates.
Mostly ZnS(Ag) scintillation detectors or proportional counters are applied (see Sect. 10.1.3 and 10.1.2)
but also other active detectors are in use.
Raabe and Wrenn have developed a method measuring the total alpha particle count number in many
successive intervals and fitting the obtained decay curve with a theoretically calculated decay function
using the maximum likelihood method [69Raa]. As a result the three activity concentrations of the radon
progeny are obtained.
A further development is the use of -spectrometry [69Mar, 80Por] or even - and -spectrometry
[97Ruz] for the identification of the different radon progeny. Silicon surface barrier or diffused junction
detectors are usually used with multichannel analysis. These methods are generally more complex,
however, with -spectrometry measurements in two intervals and with - and -spectrometry a single
measurement is sufficient. It allows also a correction for the contribution of thoron progeny. In general,
the detection limit is lower than in methods with gross alpha counting.
The spectrometry allows also a continuous measurement, if the filter and the detector are combined in
one unit. The continuous detection of -particles from 218Po and 214Po are not sufficient to determine the
concentrations of the 4 radon progeny, if no further information is available. Very often, however, an
equal ratio of the activity concentrations cPo-218/cPb-214 = cPb-214/cBi-214 is fulfilled.
Most algorithms for the calculation of concentrations from measured data assume constant progeny
concentrations during the sampling period. If this is not the case, more complex calculations are necessary
and the uncertainty will be higher.
Determination of the potential alpha energy concentration cp (PAEC)
In principle, PAEC can be calculated from measured activity concentration data of the progeny (see
Section 4.6.2). Often, however, methods are used which are simpler in instrumentation and optimised
with respect to direct PAEC determination.
A simple method is based on gross -counting during a single period [56Kus]. After a short sampling
time, a measurement of the number of -particles from 218Po and 214Po during some hours, where most of
these radionuclides decay, multiplied with the mean -energy provides a PAEC value with an uncertainty
of at least 10 % due to the difference in the -energies from 218Po and 214Po. For these measurements
mainly active detector devices (e.g. solid state detectors, electret ionisation chambers or scintillation
detectors) are used.
If shorter counting periods are used, this needs a correction of the calibration factor and an assumption
on the equilibrium status because of the different decay times of the progeny (see Fig. 10.3.1). For quasicontinuous measurements the filter and the detector must be combined in one unit. A short sampling time
may then be followed by a counting time of about 1 hour. After about 3 measurement cycles a correct
PAEC value is achieved. This may, however, not be the case, if the progeny concentrations vary strongly
in time.
Landolt-Brnstein
New Series VIII/4

Ref. p. 10-51]

10 Measuring techniques

10-49

Due to the short lifetime of 218Po (T1/2= 3.1 min) the -particle measurement needs a precise timing.
This is not the case, if instead - or -radiation from 214Pb and 214Bi is measured. Then, however,
corrections need to be applied for the influence of environmental background radiation.
While the foregoing methods provide an actual PAEC value, often a value averaged over a long time
period or the exposure Ep is wanted. In this case, in addition to the above mentioned active devices also
passive detectors, e.g. etched track and TL detectors, are used. While etched track detectors are sensitive
to -particles only, TL detectors are also sensitive to beta and -radiation, e.g. from environmental
background. In this case, two TL-detectors are applied, where one measures the background only.
The thoron progenies are also deposited on the filter. Especially the long-living progeny 212Pb
(T1/2= 10.64 h) may therefore be enriched on the filter during long sampling times and will together with
its progeny 212Bi influence the PAEC measurement. This can be checked when an additional
measurement of the filter is performed at about 10 h after the end of the sampling period, where the radon
progeny are already decayed.
Determination of the unattached fraction of the progeny fp
A separate measurement of the unattached fraction of the progeny is based on the different diffusion
properties. Due to the small size of the unattached progeny (<5 nm) compared to the aerosols they have a
much higher diffusibility and are deposited on surfaces much faster than those.
For measurements instead of a filter a diffusion battery a system of small diffusion tubes or a wire
screen is used where mainly the unattached fraction is deposited [96Por]. The further measurement of
its concentration or PAEC is similarly performed as described for the attached progeny. If a filter is
deposited after the diffusion battery, the PAEC of the attached fraction can additionally be measured.
The unattached fraction fp is then given by
fp =

cpf
c + cpf
a
p

(10.3.1.2)

where c pa is the PAEC of the progeny attached to aerosols and c pf is that of the unattached fraction.

Determination of the equilibrium equivalent concentration ce (EEC) and the equilibrium factor F.
The equilibrium equivalent concentration (EEC) ce of radon for a non-equilibrium mixture with its
progeny in air is the fictitious activity concentration of radon which is in radioactive equilibrium with its
short-lived progeny and has the same PAEC as the actual non-equilibrium mixture. The equilibrium
factor F is then defined as the quotient of ce and cRn (see Sect. 4.6.2). It is always ce cRn and hence
F 1.
ce and F are not directly measurable. A determination needs the measurement of the concentrations of
radon and their progeny. While cPo-214 can be ignored (see Sect. 4.6.2), ce of radon and F are given by the
equations
ce = 0.106 cPo-218 + 0.513 cPb-214 + 0.381 cBi-214.
F = ce / cRn
The unit of ce equals that of cRn, but is often marked Bq m3 (EEC).

Landolt-Brnstein
New Series VIII/4

(10.3.1.3)

10-50

10 Measuring techniques

[Ref. p. 10-51

10.3.1.3 Measurement of radon in the ground and in water


Radon and thoron are continuously produced in the ground by the decay of 226Ra and 224Ra, respectively.
The radon/thoron is partially transferred from minerals to pores of air in the ground (emanation) and then
it may diffuse to the surface and exhale to the atmosphere. The radon concentration in the ground air
(effective radon concentration in the ground) is about 100 to 10 000 times higher than in the open air
while for thoron the ratio is even higher. Near the surface (<1 m) the concentration decreases due to the
exhalation at the surface. The actual exhalation rate is also influenced by the air pressure and the
temperature in the atmosphere.
The radon concentration in the ground can be indirectly determined by measuring the specific activity
of 226Ra in the soil together with the determination of the emanation probability (usually 0.2 - 0.5
depending on the soil type and on the humidity of the ground). The 226Ra content in a soil sample can be
measured by -spectrometry. If the sample is deposited for about 25 days in a closed box, equilibrium of
radon with 226Ra is achieved and the total content of radon in the soil sample can also be determined by
-spectrometry (-rays from 214Pb and 214Bi).
The effective radon content in the sample can be determined when equilibrium is achieved by flowing
radon-free gas through the chamber and adsorbing the radon in an activated carbon absorber afterwards.
The radon content in the absorber is then determined by -spectrometry (-rays from 214Pb and 214Bi). The
same can be achieved by measuring the radon content in the sample before and after the flow of gas
through the chamber and taking the difference.
A direct measurement of the effective radon concentration in the ground is performed by inserting a
probe into the ground, at minimum 1 m below the surface in order to avoid surface effects. The probe
may be a special diffusion chamber for local radon measurement (see Tables 10.10 and 10.11) or a system
connected with a pump to collect a known amount of ground air into an external radon measuring device.
In this case the ground air is often returned to ground in a closed loop. In any way care is needed to avoid
atmospheric air entering the probe.
Most of the methods can also be used for the measurement of the thoron activity concentration in the
ground, if special care is taken considering the fast decay of thoron. It should also be looked at, if thoron
is influencing the radon measurement. Larger diffusion times or waiting some time before measuring a
probe may avoid these problems.
The specific activity concentration of radon dissolved in water can be directly measured in a water
sample (after 3 h when equilibrium exist between radon and its progeny) by detecting -rays from 214Pb
and 214Bi with a -spectrometer. Also a liquid scintillation detector may be used.
The radon of a water sample may also be exhaled with a radon-free gas, dried and either absorbed in a
carbon filter or transferred to a detector chamber. The radon measurement is then be performed by
methods given in Table 10.11.
A fast and simple technique which avoids the sampling of water is the use of a diffusion chamber of a
material (e.g. a membrane tube of polypropylene) which allows a fast diffusion of radon. The radon
concentration can then be conventionally measured.
Glass fibre filters of a certain brand were found to be very efficient for the adsorption of short-lived
radon decay products during filtration of water [97Phi] where they are in equilibrium with radon. The
-radiation from 214Pb and 214Bi on the dried filter can then simply be measured using a proportional
counter with a thin window.

Landolt-Brnstein
New Series VIII/4

10 Measuring techniques

10-51

10.3.1.4 References for 10.3.1


53Tsi
56Kus
57Luc
69Mar
69Raa
72Tho
80Bug
80Por

81Sco
84Geo
84Naz
88Naz
88Kot
88NC
90Geo
92Bal
96Geo
96Por
97Phi
97Ruz
98NN
00How
02SSK

Tsivoglou, E.C., Ayer, H.E., Haladay, D.A.: Occurrence of non equilibrium atmospheric
mixtures of radon and its daughters. Nucleonics 11(9) (1953) 40.
Kusnetz, H.L.: Radon daughters in mine atmospheres. A field method for determining
concentrations. Am. Ind. Hyg. Assoc. J. 17 (1956) 85.
Lucas, H.F.: Improved low-level alpha scintillation counter for radon. Rev. Sci. Inst. 28
(1957) 680.
Martz, D.E., Hollemann, D.F., McCurdy, D.F., Schiager, K.J.: Analysis of atmospheric
concentrations of RaA, RaB and RaC by alpha spectroscopy. Health Phys. 17 (1969) 131.
Raabe, O.G., Wrenn, M.E.: Analysis of the activity of radon daughter samples by weighted
least squares. Health Phys. 17 (1969) 593.
Thomas, J.W.: Measurement of radon daughters in air. Health Phys. 23 (1972) 783.
Bugsin, A., Phillips, C.R.: Uncertainties in the measurement of airborne radon daughters.
Health Phys. 39 (1980) 943.
Porstendrfer, J., Wicke, A., Schraub, A.: Methods for a continuous registration of radon,
thoron and their decay products indoors and outdoors, in: Gesell, T.F., Lowder, W.M. (eds).
Natural radiation Environment III. CONF-780422 Vol. 2, DOE, Washington D.C., 1980,
p. 1293-1307
Scott, A.G.: A field method for measurement of radon daughters in air. Health Phys. 41
(1981) 403.
George, A.C.: Passive integrated measurement of indoor radon using activated carbon. Health
Phys. 46 (1984) 867.
Nazaroff, W.W.: Optimizing the total alpha three count technique for measuring
concentrations of radon progeny in residences. Health Phys. 46 (1984) 395.
Nazaroff, W.W., Nero, A.V.: Radon and its decay products in indoor air. New York,
Chichester, Brisbane, Toronto, Singapore: John Wiley & Sons, 1988.
Kotrappa, P., Dempsey, J.C., Hickey, J.R., Stieff, L.R.: An electret passive environmental
222
Rn monitor based on ionization measurement. Health Phys. 54 (1988) 47.
National Council on Radiation Protection and Measurement: Measurement of Radon and
Radon Daughters in Air. NCRP Report No. 97, Bethesda, MD, 1988.
George, A.C., Webber, T.: An improved passive activated carbon collector for measuring
environmental 222Rn in indoor air. Health Phys. 58 (1990) 583.
Baltzer, P., Gorsten, K.G., Backlin, A.A.: A pulse counting ionization chamber for measuring
the radon concentration in air. Nucl. Inst. Meth. Phys. Res. A317 (1992) 357.
George, A.C.: State-of-the-art instruments for measuring radon/thoron and their progeny in
dwellings a review. Health Phys. 70 (1996) 277.
Porstendrfer, J.: Radon: measurements related to dose. Environ. Int. 22, Suppl. 1 (1996)
563.
Philipsborn, H. von: Efficient adsorption of waterborn short-lived radon decay products by
glas fiber filters. Health Phys. 72 (1997) 451.
Ruzer, L., Sextro, R.: Measurement of radon decay products in air by alpha and beta
spectrometry. Radiat. Prot. Dosim. 72 (1997) 43.
NNDC: Nuclear Data, Decay Radiations. National Nuclear Data Center, Brookhaven
National Laboratory, Upton, NY, 1998.
Howarth, C.B., Miles, J.C.H.: Results of the 1998 European Commission intercomparison of
passive radon detectors. European Commission, Report EUR 18835 EN, Luxembourg, 2000.
Strahlenschutzkommission: Leitfaden zur Messung von Radon, Thoron und ihren
Zerfallsprodukten. Verffentlichungen der SSK, Band 47, Mnchen, Jena: Urban & Fischer,
2002.

Landolt-Brnstein
New Series VIII/4

10-52

10 Measuring techniques

[Ref. p. 10-81

10.3.2 In vivo measurements


10.3.2.1 Introduction
Internal exposures due to incorporation of radioactive materials may be detected both by in vivo and in
vitro measurement: the in vivo measurement involves the measurement by detectors external to the body,
thus mainly radionuclides emitting or X-rays can be detected by this way. The in vitro method is based
on the measurement of the activity excreted with the urine, faeces and exhaled air and thus can be applied
in principle for any material. In many cases the in vitro method is more sensitive but the interpretation of
the results is in general more difficult because of the lack of information with respect to the individual
metabolic behaviour of the incorporated materials. The in vivo method is not as sensitive as the in vitro
method but the evaluation of the results is more easy because not that much information is needed for the
assessment of dose from body or organ activity data. So the in vivo measurement is considered to be the
best method for the detection of emitting radionuclides i.e. most of the fission and activation products
and few actinides such as 235U or transuranium radionuclides such as 241Am, whereas the in vitro method
is applied for the detection of all other radionuclides where the in vivo method is not sensitive enough. In
few cases the in vivo technique can be applied also for -emitters with emitting daughters, such as 226Ra
or 238U.
The first in vivo measurements of Radium have been carried out in 1927 by Blumgart and Weiss using
ionisation chambers for blood flow studies [27Blu]. The lower detection limit of those measurements was
reported to vary in the range from 5 to 100 g (0.18 to 3.7 MBq) 226Ra, assuming radiological equilibrium
between 226Ra and its -emitting daughter products [29Sch]. In 1931 Schlundt took into account the
geometry and the self-absorption of the body by inserting sealed radon sources in a phantom, thus
providing the first calibration standard for in vivo measurements [31Sch]. In those years three lethal cases
of radium poisoning were established in the radium manufacturing industry in Germany, this enforcing
the further development of detection procedures at the Max-Planck Institute for Biophysics in Frankfurt,
Germany. So in 1938 the Untersuchungsstelle fr die physikalische Diagnostik der Radiumvergiftungen was founded at the Kaiser-Wilhelm-Institut for Biophysics in Frankfurt. At the same time
researchers in the United States applied high sensitive Geiger-Mller tubes also and the lower detection
limit was reported to be 1 g (37 kBq) 226Ra [37Eva]. Few years later Rajewsky and Dreblow performed
first partial body measurements using a so-called gamma-ray stethoscope which enabled the diagnosing
physician to contact directly the individual parts of the body thus allowing for the localization of radium
deposits in the body [41Raj]. About one decade later, Sievert developed a 4 - geometry whole-body
counter using an arrangement of 10 long ionisation chambers surrounding the subject. For achieving a
low background, the measuring device was installed into granite rocks 50 m below ground surface, this
resulting in a detection limit of about 1 nCi (37 Bq) of 226Ra for a measurement of 3 to 4 hours [51Sie,
57Sie]. In the fifties a lot of progress was achieved due to the development of new types of detectors such
as NaI(Tl) scintillation detectors as well as liquid scintillation detectors [57And] and organic scintillation
detectors [58Bir]. In 1956 the first technical conference on in vivo measurement was organised in Leeds
in order to discuss the state of the art of measuring techniques. Since that time further improvements in
the sensitivity and reliability of in vivo measurements have been achieved by the use of arrays of
scintillation detectors and the application of anticoincidence techniques for the reduction of the detectors'
background. In 1968 Laurer presented the first dual NaI(Tl)/Cs(Tl) scintillation crystal detector
(phoswich) which was designed especially for low energy photon detection [68Lau]. In the late sixties
semi-conductor detectors were introduced for in vivo measurement, starting with Lithium-drifted
Germanium (Ge(Li)) detectors, which later on have been replaced by high purity Germanium (HPGe)
detectors.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-81]

10 Measuring techniques

10-53

With improving measuring techniques, whole-body counting has become a standard method for
radiation protection. Personnel working in nuclear installations or in laboratories handling radioactive
materials may receive internal radioactive contamination by inhalation, ingestion or intake through wounds.
Techniques of in vivo measurement may be employed in the monitoring and control of such contamination,
either as the sole means of evaluation or in conjunction with in vitro measurement of activity excreted by the
subject or present in the working environment.
10.3.2.2 Requirements
Techniques of in vivo measurement depend on the detection outside the body of photons originating from
internally deposited radioactive materials. They are useful, as a means of controlling exposure to a given
nuclide, if a significant deposit of that nuclide leads to a detectable signal at or near the surface of the body. In
this context, a significant deposit would be one implying an intake at or exceeding some level requiring
administrative action. Their feasibility in the case of a given nuclide thus depends on the yield and energy of
photons emitted by the nuclide, on its pattern of deposition in the body and on the relevant limits on internal
exposure. Many fission and activation products emit abundant penetrating gamma radiation, thus allowing for
the assessment of intakes small in relation to annual limits, with relatively simple equipment. By contrast,
long-lived -emitters with only weak low-energy photon emissions may escape detection with the most
sensitive and elaborate equipment, even when present in levels far exceeding annual limits on intake; in such
instances the technique will find application only in the investigations of major acute intakes or in the
monitoring of uptake following long-term chronic exposure.
In most applications, photon detectors are located at selected sites near or on the body. Usually, at least
partial shielding of the detector and/or of the subject will be needed to reduce the interfering response from
ambient radiation; in some cases anticoincidence techniques may be required to achieve sufficient background discrimination. Electrical signals from the detectors must be amplified and processed, leading to a
gamma-ray spectrum which will most conveniently be stored in computer based systems. Procedures are
necessary to separate the response attributable to a given nuclide in the body from that due to ambient
radiation and to components associated with other sources of body radioactivity. The extracted response must
be converted into an assessment of body or organ radioactivity through appropriate calibration procedures.
Three detection features define the requirements for in vivo measurement of radionuclides in the human
body:
1. Selectivity: the capability to measure the activity of a radionuclide in the presence of other
radionuclides.
2. Sensitivity: the response of the measuring system with respect to the level of radioactivity within the
body, i.e. the capability to measure internal exposures below the limits.
3. Accuracy: the mean deviation of the result in terms of radioactivity from the actual radioactivity in the
body or in a phantom, respectively.
Selectivity
The monitoring method has to be prepared to provide nuclide specific information, which is basic
requirement for internal dose assessment. Except for circumstances where there is only one radionuclide
handled by the workers to be monitored, the measurement system should provide identification of
radionuclides. This means that in case of direct monitoring and also when indirect methods are applied for
measuring gamma emitting radionuclides, gamma spectrometry is the method, which can meet this
requirement. In this respect the semiconductor spectrometry provides better selectivity compared to
scintillation spectrometry.
Sensitivity
The monitoring sensitivity should be high enough to be able to determine with proper safety an internal
exposure corresponding to 1 mSv annual effective dose or 10 % of organ dose limits, respectively. To
meet this requirement one has to consider the minimum detectable activity for the radionuclides to be
expected, the selected monitoring frequency and a reasonable measuring time. This is illustrated by
Landolt-Brnstein
New Series VIII/4

10-54

10 Measuring techniques

[Ref. p. 10-81

Tables 10.12 and 10.13 which show the characteristics for some selected fission and activation products
and some selected actinides, respectively. Both the energy and the yield of the predominant photon
radiation of most of the fission and activation products are relative high. On the other hand the dose
coefficients of these radionuclides are relatively low. So the intake corresponding to 1 mSv is rather high
and thus the respective body or organ activities are high even at the end of long monitoring intervals.
Contrary, the energy and the yield of the photon radiation of the actinides are typically low and the dose
coefficients are high as compared to the fission and activation products.
Table 10.12. Radiological and monitoring characteristics for selected fission and activation products
[83ICR, 94ICR, 04BMU]
Nuclide
22

Na

57

Co

60

Co

125

131

134

Cs

137

Cs

Predominant photon radiation Typical


absorption
Energy [keV] Yield [%]
type
511
180
F
1275
100
122
86
M
136
11
1173
100
M
1333
100
40
F
27.2 (K2)
74
27.5 (K1)
14
31.0 (K1)
6.7
35.5
365
82
F

Routine monitoring
Intake1) corresponding to 1 mSv Interval [d]
Required seneff. dose [Bq]
sitivity [Bq]
180
200
5 105
(Whole body)
180
20000
2.6 106
(Whole body)
180
1000
1.4 105
(Whole body)
120
700
1.4 105
(Thyroid)

9.1 104

14

569
605
796
662

1 105

180

1.5 105

180

15
98
85
85

100
(Thyroid)
6000
(Whole body)
10000
(Whole body)

1) Inhalation of aerosols with 5 m AMAD particle size

Table 10.13. Radiological and monitoring characteristics for selected actinides [83ICR, 94ICR, 04BMU]
Predominant photon radiation Typical
Routine monitoring
Nuclide
Intake1) corresabsorption ponding to 1 mSv Interval [d]
Required senEnergy [keV] Yield [%]
type
eff. dose [Bq]
sitivity [Bq]
235
U
145
11
M
550
180
3
186
57
(Lungs)
239
Pu
1.6
S
120
180
2
13.6 (L)
2.3
(Lungs)
17.1(L)
0.6
20.3 (L)
241
13.6
M
37
180
0.2
Am
13.9 (L)
18.6
(Lungs)
17.6(L)
0.3
20.3 (L)
35.9
(Skeleton)
59.6
1) Inhalation of aerosols with 5 m AMAD particle size

The range of the photons in the body governs the sensitivity of in vivo measurement. For soft tissue
the range of the 17.1 keV X-rays of 239Pu is 0.89 cm, and so most of the photons emitted from an internal
contamination are absorbed within the body. The range of the 1332 keV -rays of 60Co, however, is
16.5 cm and so most of the photons emitted in the body will reach the body surface. Thus, the photon flux
Landolt-Brnstein
New Series VIII/4

Ref. p. 10-81]

10 Measuring techniques

10-55

at the body surface due to a 60Co deposition will be at least 3 orders of magnitude higher than that due to
a 239Pu deposition with the same activity. Moreover, when taking into account the different dose
coefficients, the photon flux due to an internal 60Co exposure will be more than 6 orders of magnitude
higher than that due to a 239Pu exposure with the same committed effective dose, this illustrating the range
of sensitivity required for in vivo measurements.
Accuracy
In monitoring of occupationally exposed workers for radiation protection purposes, procedures must be
established to ensure that workers have exposures measured and recorded with a reasonable degree of
accuracy. General requirements for the overall accuracy of the dose assessment have been recommended
by the ICRP [97ICR]. Special requirements for in vivo measurement have been defined for example by
the U.S. Department of Energy in its Laboratory Accreditation Program DOELAP [99USD]. These
recommendations have been adopted by national guidelines, as for example the German Guideline for
Internal Monitoring [04BMU]. The various national and international guidelines have been harmonised in
the framework of the European project IDEAS [03DOE].
The accuracy of a monitoring result reflects to the quality of measurements and is usually
characterized by two quantities namely by the bias and precision (repeatability).
The relative bias (Br) is a measure of how close the assessed activity is to the actual activity in the
organ(s) or in the whole body. Since the actual activity in the person is rarely known, this criterion applies
to measurements on suitable phantoms that simulate the person. The relative bias statistic (Bri) is defined
for the purposes of performance testing of a finite number of measurements in each category of analysis
by
Br =

1
N

ri

i =1

with
Bri =

Ai Aai
Aai

(10.3.2.1)

where
N is the number of test measurements in a given category (N 5)
Ai is the value of the i th measurement in the category being tested
Aai is the actual quantity in the test mock-ups (phantom) for the ith measurement
For service laboratories the performance requirement for the relative bias should be 0.25 Br 0.50.
This requirement can only be considered if all values of Aai exceed the lower limit of detection by at least
factor 5.
The relative precision (SB) describes the relative dispersion of the values of Bri from their mean Br and
is defined as
N

(B

ri

SB =

i =1

Br ) 2

N 1

(10.3.2.2)

For service laboratories the relative precision should be SB 0.4 for the conditions mentioned above,
i.e. if all Aai exceed the lower limit of detection significantly.

Landolt-Brnstein
New Series VIII/4

10-56

10 Measuring techniques

[Ref. p. 10-81

10.3.2.3 Principles of spectrometry


Photon interactions
For understanding spectrometry a knowledge of the basic processes by which a photon interacts with
matter is essential. Three fundamental processes govern the interaction: photoelectric effect, Compton
scattering and pair production. Due to these interactions the intensity I(x ) of the photon flux is decreasing
along the pathway x according to the function
I ( x) = I 0 e x

(10.3.2.3)

where is the total linear attenuation coefficient. For illustration Fig. 10.33 shows the linear attenuation
coefficient of water and NaI, respectively. Water is a typical example for a low-Z material, the absorption
behaviour of which is similar to soft tissue, and NaI is a typical example for a high-Z material, which is
widely used for spectrometry. More detailed information about photon interaction can be found in
Section 3.5.2.3 and elsewhere [80Tai, 99Kno].
10 2

10 2

H2 O

5
-1

10

Linear attentuation coefficients [cm

Linear attentuation coefficients [cm

-1

5
2

Total

5
2

10-1
5
2

Compton

10-2

Photoelectric

5
2

10-3
10-2

10
2
5
Energy [MeV ]

10
5
2

1
5
2

10-1

Total

Compton

10-2
5

Photoelectric

Pair
production

Pair production
-1

Nal

10

10-3 -2
10

10-1 2
5
1
Energy [MeV ]

10

Fig. 10.33. Linear attenuation coefficient of H2O and NaI.

The photoelectric capture predominates for low photon energies and the photons are absorbed much
more strongly in high-Z materials than in low-Z materials. If the incident photon is absorbed by
photoelectric effect in a detector, the resulting pulse contributes to the so-called full absorption peak or
photo-peak, which provides the key information about the energy and the intensity of the incident photon
radiation. If the photon undergoes Compton scattering, the resulting pulse contributes to the so-called
Compton continuum of the detector spectrum which consists of two components: if the Compton
scattering occurs inside the detector the resulting pulse contributes to the detector-specific Compton
continuum which covers according to the energy of the scattered electron the energy range from zero for
= 0 up to the so-called Compton edge for = 180. If on the other hand the Compton scattering occurs
in the environment and the scattered photon is subsequently absorbed in the detector, then the resulting
pulse contributes to the environment-specific Compton continuum which covers the energy range from
the so-called backscatter-peak for = 180 up to the photo-peak for = 0. It is important to know that
anticoincidence techniques can reduce only the detector-specific Compton continuum but not the
environment-specific Compton continuum. More detailed information about photon interactions is given
in Section 3.5.2.3.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-81]

10 Measuring techniques

10-57

Energy resolution
The energy resolution of a detector describes its ability to distinguish between photon energies. When
exposed to a radioactive source, the detection system processes a number of monoenergetic photons,
which results in a spectrum consisting of the photo peak and the Compton continua. For illustration
Fig. 10.34 and Fig. 10.35 show the spectra of a NaI(Tl) scintillation detector (see Section 10.3.2.4.4.1) for
low-energy photons (5.9 keV K X-rays from 55Fe) and for high-energy photons (662 keV -rays from
137
Cs), respectively. The low-energy photons are fully absorbed in the detector because at 5.9 keV the
probability for photoelectric absorption is more than two orders of magnitude higher than the probability
for Compton scattering (Fig. 10.33). On the other hand, the high energy photons undergo mainly
Compton scattering in the detector because at 662 keV the probability for photoelectric absorption is one
order of magnitude less than that for Compton scattering. Thus, in the spectrum for low-energy photons
there is only the photo peak whereas in the spectrum of high-energy photons there is in addition a broad
Compton continuum including the backscatter peak and the Compton edge. Ideally, all fully absorbed
monoenergetic photons would be assigned exactly the same pulse height (or channel) in the measured
spectrum. However, the photo-peak in the measured spectrum is a distribution of pulse heights with a
peak width that reflects the detector resolution. The full width at half maximum (FWHM) of the photopeak is used to characterise the resolution of the detector. The FWHM is the energy width of the
distribution at half the maximum of the photo-peak when the background has been subtracted. For lowresolution detectors, such as scintillation detectors, resolution is defined as the FWHM divided by the
photo-peak energy, and is usually expressed as a percentage. The 662 keV gamma ray from 137Cs is
usually used as the reference for this purpose.
For high-resolution semiconductor detectors, the energy resolution is usually specified as the FWHM
(keV or eV) for a specified energy. Manufacturers normally also provide the width of the photo-peak at
one-tenth and one-fiftieth of the maximum, with reference values for the 1.332 MeV gamma ray from
60
Co or the 122 keV gamma ray from 57Co, depending on the type of detector.
70000

1400
59

Photomultiplier
1200 tube noise

600
FWHM
77 channels

Peak channel = 70
Resolution =6/70 = 8.6%

40000
30000
20000

FWHM
6 channels
Backscatter
peak

10000

200
0

50000

Counts per channel

800

400

50

100 150 200 250


Channel number

300

350

Cs source

60000

Peak channel = 177


Resolution = 77/177 = 43.5%

1000

Counts per channel

137

Fe source

400

Fig. 10.34. Spectrum of a NaI(Tl) scintillation


detector for low-energy photons (5.9 keV K X-rays
from 55Fe).

20

Comption
edge
60
40
Channel number

80

100

Fig. 10.35. Spectrum of a NaI(Tl) scintillation detector


for high-energy photons (662 keV -rays from 137Cs).

Detection efficiency
The detection efficiency is defined as the ratio of the number of photons detected to the number of
photons emitted by a radiation source during a given time interval. The detection efficiency is made up by
four factors:

Landolt-Brnstein
New Series VIII/4

10-58

10 Measuring techniques

[Ref. p. 10-81

1. Geometrical attenuation factor: the fraction of all emitted photons which are emitted in the direction
of the sensitive volume of the detector, depending on the solid angle covered by the detector with
respect to the source.
2. Material attenuation factor: the fraction of those photons emitted in the direction of the sensitive
volume, which actually reach it, depending on the attenuation of the material in between the source
and the detector.
3. Interaction efficiency: the fraction of photons reaching the sensitive volume that react with it,
depending on the attenuation of the detector material.
4. Data recording efficiency: the fraction of photons interacting with the sensitive volume, which
produce recorded events, depending on the type of recording (i.e. photo-peak counting or total
counting) and the data acquisition system.
10.3.2.4 Equipment
10.3.2.4.1 Detectors
10.3.2.4.1.1 Scintillation detectors (see also Section 10.1.3)
There are three groups of scintillation detectors: crystals, glasses and gases. For in vivo measurements,
however, only crystal type scintillation detectors are applied. Each photon, which interacts with the
sensitive volume of the crystal, generates a single scintillation pulse. This is a very weak pulse, typically
consisting of less than 1000 photons of few eV, so it has to be viewed by a highly sensitive photomultiplier tube (PMT) and the whole assembly must be enclosed in a light-tight housing to isolate the
scintillation from the ambient light. The characteristic properties of some selected scintillation materials
are summarized in Table 10.14.
Table 10.14. Properties of NaI(Tl), CsI(Tl), Bi4Ge3O12, and organic (Polystyrenetetraphenylbuta-diene)
scintillators.
Property
Density [g/cm3]
Light output relative to NaI(Tl)
Wavelength of maximum emission [nm]
Decay constant [s]
Hygroscopic
Energy resolution at 662 keV [FWHM in %]

NaI(Tl)
3.67
1
415
0.23
Yes
7 - 10

CsI(Tl)
4.51
0.45
550
1.0
No
10 - 12

Bi4Ge3O12
7.13
0.12 - 0.20
480
0.3
No
10 - 12

Organic
1.0
0.14
450
0.005
No
25 - 50

Sodium iodide activated with thallium (Nal(Tl)) provides the best properties with respect to light
output, decay constant and energy resolution. However, NaI(Tl) is hygroscopic, and absorption of water
results in a loss of energy resolution. So these crystals must be placed in gas-tight housings. A typical
design of a NaI(Tl) scintillation detector is shown in Fig. 10.36. In between the crystal and the housing
there is a MgO reflector and so the scintillation light can leave the crystal only via the optical window.
Thus most of the scintillation light is collected on the photo cathode of the photomultiplier tube behind
the optical window. A magnetic shield protects the photomultiplier tube in order to avoid deflection of the
secondary electrons in the tube by external magnetic fields. Both the reflector and the magnetic shield
make sure that the amplitude of the output signal is proportional to the energy absorbed in the NaI(Tl)
crystal.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-81]

10 Measuring techniques

10-59

62

Magnetic shield

187
216

130 mm Photo tube


Optical window

25

Mounting holes - 6 M6 on 200 B.C.


Reflector - packed MgO

H + 13

NaI (Tl) crystal


Spun body - 0.8 Al 2S
184.2

Fig. 10.36. Typical design of


a NaI(Tl) scintillation detector
(Harshaw matched window
assembly with crystal dimensions: 178 mm ; 130 mm
height).

Caesium iodide activated with thallium (CsI(Tl)) can also be used as scintillator material, but the light
output is smaller and thus the energy resolution is not as good as that of NaI(Tl). So CsI(Tl) crystals are
not commonly used for spectrometric measurements but they are frequently used as anticoincidence
detectors in order to reduce the background of NaI(Tl) crystals, this requiring high efficiency but no highenergy resolution. The combination of NaI(Tl) and CsI(Tl) crystals as a dual phosphor sandwich
(phoswich) allows for high sensitive detection of low-energy photons (see Section 10.3.2.4.3.2).
Because of the poor energy resolution of the scintillation detectors the photo peaks are relatively
broad and thus the background prediction can be difficult, especially in the low-energy region. This is
illustrated by Fig. 10.37 which shows the spectrum of a NaI(Tl)/CsI(Tl) phoswich detector for a subject
with 0.25 kBq 241Am in the liver, 1.5 kBq 241Am in the skeleton and 12 kBq 137Cs in the whole body. The
photo peak due to the X-rays of 137mBa (daughter of 137Cs) is overlapping to some extend the photo peak
due to the -rays of 241Am, and thus the separation of the photo peaks from each other and from the
Compton continuum requires high sophisticated spectrum evaluation procedures (see Section 10.3.2.5).
4000
3500

Counts per channel

3000

241
137m

Am:59.6 keV

Ba:31.8 /32.2 keV

2500
2000

Fig. 10.37. Spectrum of a NaI(Tl)/CsI(Tl) phoswich


detector (Harshaw 208 mm matched window
assembly with 1 mm thick NaI(Tl) crystal and 51 mm
thick CsI(Tl) crystal) for a subject with 0.25 kBq 241Am
in the liver, 1.5 kBq 241Am in the skeleton and 12 kBq
137
Cs in the whole body (detector arranged over the
liver, measuring time 2000 s).

1500
1000
500
0

Landolt-Brnstein
New Series VIII/4

20

60
40
Channel number

80

100

10-60

10 Measuring techniques

[Ref. p. 10-81

Bismuth germanate (Bi4Ge3012), often abbreviated to BGO, provides the highest efficiency of all
available scintillation materials because of its high density and effective atomic number. The energy
resolution, however, is relative poor and so BGO is applied only in those cases where small detectors with
high photo peak efficiency are required, as for example wound measurements.
Organic scintillation detectors
Solutions of organic liquids [61Lan], and solid organic scintillators [62Bur], have also been used for in
vivo measurement applications. Solid organic scintillators can be made by impregnating plastic materials
with anthracene. They can be made in very large sizes (e.g. 60 40 10 cm3) but require several
photomultiplier tubes to achieve even a modest energy resolution, and in consequence have not been widely
adopted for radiological protection purposes. They could be considered only where the interest was in a single
nuclide, or in a mixture whose composition was reliably known, and where interference from the body's
natural 40K could be either neglected or inferred from measurements prior to the subject's exposure. Organic
scintillators can also be incorporated into liquid solvents; geometries approaching 4 can be produced with
such solutions contained in annular tanks, but they suffer from the same restrictions as organic scintillators.
Organic scintillation detectors have almost the same photon absorption behaviour as soft tissue and
thus they can be applied for direct assessment of the actual internal dose rate due to incorporated
-emitting radionuclides [95Doe].

10.3.2.4.1.2 Semiconductor detectors


Semiconductor detectors are solid-state ionisation chambers, the principle of which being described in
detail in Section 10.1.4 and elsewhere [92Del, 99Kno, 80Tai]. For in vivo measurements most commonly
Germanium detectors are used, starting in former times with Li drifted Germanium detectors (Ge(Li)),
which have been replaced since 1976 by high purity Germanium detectors (HPGe) [76Fal]. In the eighties
mainly p-type HPGe crystals have been used. These crystals have a lithium diffusion zone to form the
n-contact, which results in an insensitive layer of about 0.6 mm at the crystal surface. So only photons
with energy higher than about 50 keV could be detected. Since the nineties also n-type detectors are been
applied which have a boron ion implantation to form the p-contact. The implantation results in a very thin
insensitive layer (0.3 m) at the front of the crystal, thus allowing also for measurement of low-energy
photons with energies of several keV.
Semiconductor detectors have major advantages in energy resolution, the FWHM being typically
below 0.6 keV for low energy photons or 2 keV for high energy photons, respectively (Table 10.15).
Thus, semiconductor detectors allow almost unambiguous identification of the radionuclides in a mixture,
but most of them are inconvenient in that they need cooling to liquid nitrogen temperatures. High purity
germanium (HPGe) detectors can tolerate cycles to room temperature but need cooling during operation.
Furthermore, many semiconductor detectors are available only in fairly small sizes, so that their
geometrical efficiency is small as compared to inorganic crystals and other scintillators. Compact arrays
of three to six detectors are becoming standard for monitoring contamination in specific organs, such as
the lungs.
Miniature semiconductor detectors, in particular those using cadmium telluride (CdTe) operating at
room temperatures, are becoming increasingly available. CdTe detectors offer high sensitivity for
detection of low energy photons. Their small size (approximately 10 mm in diameter and 2 mm thick)
makes them ideal for localized wound monitoring.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-81]

10 Measuring techniques

10-61

Table 10.15. Properties of some selected semiconductor detector materials.


Property
Density [g/cm3]
Band gap at 300 K [eV]
Energy per electron-hole pair at 77 K [eV]
Requires cooling
Energy resolution at 5.9 keV [FWHM in keV]
Energy resolution at 1332 keV [FWHM in keV]

Si
2.33
1.12
3.61
Yes1)

Ge
5.33
0.67
2.98
Yes
0.6
1.8

CdTe
6.06
1.47
4.43
No

1) If high resolution is required

The excellent energy resolution of HPGe detectors is illustrated by Fig. 10.38., which shows the
spectrum for a subject with 0.25 kBq 241Am in the liver, 1.5 kBq 241Am in the skeleton and 12 kBq 137Cs
in the whole body. A comparison of this spectrum with the corresponding phoswich spectrum measured
at the same subject in the same geometry at almost the same time (Fig. 10.37) reveals the major
advantage of the HPGe detectors especially for the in vivo measurement of low-energy photon emitters.
200
180
241

Counts per channel

160

Am:59.6 keV

140
120

137m

Ba:31.8 /32.2 keV

100
80
60
40
20
0

200

400
600
Channel number

800

1000

Fig. 10.38. Spectrum of a HPGe detector (Silena 50 %


coaxial n-type HPGe crystal) for a subject with 0.25 kBq
241
Am in the liver, 1.5 kBq 241Am in the skeleton and 12
kBq 137Cs in the whole body (detector arranged over the
liver, measuring time 2000 s).

10.3.2.4.1.3 Gas-filled detectors


There are three types of gas-filled detectors used for radiation protection measurements: Geiger-Mller tubes
for counting of absorption events, ionisation chambers for dosimetry, and proportional counters for
spectrometry. Large-area proportional counters with anticoincidence guard layers offer energy resolutions
intermediate between those of scintillation counters and semiconductor detectors and, with acceptable
detection efficiency at energies below 30 keV, they were in former times seen as the most profitable approach
to the assessment of plutonium in lungs. Several designs were produced by individual laboratories using
argon-methane or xenon-methane counting gas at normal or high pressure. The energy resolution was
13-15 % and the absorption probability was 30-85 % for the 13.6 and 17.2 keV uranium L X-rays from
the decay of 239Pu. High sophisticated designs were developed using full space anti-coincidence
techniques for background suppression [76Sch]. However, the proportional counters fell into disuse with
the development of the phoswich, which offered greater sensitivity and robustness.

Landolt-Brnstein
New Series VIII/4

10-62

10 Measuring techniques

[Ref. p. 10-81

10.3.2.4.2 Electronics (see also Section 10.1.3)


Electronic equipment is required to extract, amplify and sort electrical signals from the detector, converting
them ultimately into a pulse-height distribution. It therefore includes a chain of preamplifier and main
amplifier for pulse shaping and amplification followed by an analogue-to-digital converter and multi-channel
analyzer for pulse height analysis; besides, units with specialized functions are required for phoswich
detectors, or for controlling the movement of detectors scanning, high voltage power supplies, etc.
Preamplifier
The preamplifier being located close to the detector serves as a gain stabiliser and an impedance matcher. For
scintillation detectors mainly voltage-sensitive preamplifier is used whereas for semiconductor detectors
charge-sensitive preamplifiers are preferred. The preamplifier adds no or little amplification to the signal but
it enables the signal to pass without loss of information through a coaxial cable to the main amplifier, which
in general is located out of the shielding in several meters distance from the detector.
Main amplifier
The main amplifier serves for amplification and proper shaping of the pulses from the preamplifier. Modern
amplifiers provide many functions for smoothing and shaping of the pulses by integration and differentiation,
baseline restoration, pole-zero adjustment and linear amplification, the gain varying from a factor 10 to 5000.
The output signal can be chosen to be unipolar (either positive or negative) or bipolar (first positive and then
negative), the standard for spectroscopic applications being a positive unipolar pulse with amplitude up to
10 V. The length of the pulses is controlled by the shaping time constants and should not exceed 10 s in
order to avoid superposition of pulses at higher count rates which would result in broadening of the photo
peaks and thus in a loss of energy resolution of the detector system.
Pulse-height analysis
For determination of the amplitude the pulses are first digitised by an analogue-to-digital converter (ADC)
and then fed into a multichannel analyser (MCA). The MCA reads the pulses according to their height into a
memory consisting of a certain number of channels, thus generating a spectrum i.e. a frequency distribution
of the pulses as a function of their amplitude. The total number of channels per spectrum is defined by the
width of the photo peaks. It should be large enough to provide full information about the peak shape in order
to allow for proper separation of the photo peak from the background continuum or from overlapping photo
peaks due to other radionuclides, respectively. On the other hand the number of channels should not be too
large in order to have a sufficient number of pulses per channel or good counting statistics, respectively. For
scintillation detectors 0.25k (256) channels are sufficient to cover the energy range from zero to 3000 keV,
whereas for HPGE detectors up to 8k (8096) channels may be required to provide full information in this
energy range.
At present time, both ADCs and MCAs are available as plug-in components of personal computers
and customized computer codes for the qualitative and quantitative analysis of the measured spectra are
available for all gamma spectrometry systems. The computer codes include basic functions such as
background subtraction, and also more ambitious operations such as adjustment for instrumental drift,
resolution of a spectrum into its several components by linear regression analysis and peak search, evaluation
and identification procedures (see Section 10.3.2.5).
10.3.2.4.3 Shielding
The purpose of shielding is first of all to reduce the background radiation to the level necessary for the
sensitivity required; but also to reduce perturbations in the counter background response, which occur because
the subject's body distorts the ambient radiation field through absorption, scattering and other processes. The
background is governed by cosmic radiation and the radiation of radioactive materials present in the local
environment (see Chapter 11). The cosmic radiation consists primarily of charged particles of solar or
galactic origin, which produce mesons, electrons, photons and activation products such as 7Be or 14N and

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-81]

10 Measuring techniques

10-63

16

O due to interaction with the earth atmosphere. The radioactivity in the local environment is mainly due
to natural uranium and 40K, but also due to man-made 137Cs from the nuclear weapons fallout and the
Chernobyl accident, respectively. Also airborne activity such as 222Rn and 220Rn progeny may contribute
significantly to the background radiation in the environment of the detector device. In addition, radionuclides in the detector device itself may give rise for some background components, i.e. 60Co in steel
components, uranium in aluminium and beryllium components, and 40K in photomultiplier tubes.
The background may be characterized by the background index that gives the count rate per unit
detector volume, typically in units of counts per minute per cm3 over the energy range from 200 keV to
2 MeV. The reduction of the background is characterized by the background reduction factor indicating
the ratio of count rates measured with a detector in a well defined energy range without and with applying
a certain procedure for background reduction, as for example outside and inside a shielding room.
In principle, there are passive and active methods for background reduction: the passive methods are
based on the absorption of environmental radiation with appropriate shielding materials and/or on the
selection of construction materials with very low intrinsic radioactivity in the environment of the detector
device whereas the active methods are making use of special anticoincidence techniques for reducing the
detector specific background signal.
10.3.2.4.3.1 Passive shielding
The requirements for primary shielding materials are: high attenuation of gamma rays, requiring high atomic
number and density; freedom from unacceptable concentrations of natural or artificial radionuclides; and
suitable mechanical properties for fabrication and assembly. Steel or lead are most commonly used. If lead is
chosen, there will be characteristic X-rays, induced by ambient radiation or by the subject's gamma-ray
emissions; if these interfere in a critical energy region, they may be removed through an inner lining of a few
mm of cadmium or tin, with the ensuing Cd or Sn X-rays eliminated if necessary by a further lining of steel or
copper. For major installations typical thicknesses for the primary material are 5-10 cm lead or 10-20 cm
steel. Those thicknesses seem to be optimum due to the fact that the background from the environment is
about two orders of magnitude higher than the background from the subject due to the natural 40K in the body.
So it is making no sense to reduce the environmental background far below the background from the subject.
Thus, a background reduction factor of 100 seems to be reasonable with respect to in vivo measurements in
the energy range up to about 3000 keV. Such a background reduction factor is achieved for example by
14.6 cm steel (Table 10.16). Behind the shielding the environmental radiation is scattered down to a broad
Compton continuum with a maximum around 200 keV. The Compton continuum may be attenuated by a
factor of 100 with an inner lining of about 5 mm Pb. In the Pb lining, however, characteristic X-rays are
produced with energy between 72 and 88 keV (K1 at about 75 keV). These X-rays then may be absorbed
by an additional inner lining of about 2 mm Sn, and the X-rays from the Sn (K1 at about 25 keV) then
may be absorbed by a third inner lining of about 0.25 mm Cu, this being the principle of the so-called
graded Z lining which provide optimum shielding for in vivo measurements (Table 10.16).
Table 10.16. Design parameters for a graded Z shielding.
Critical radiation

Reference
energy [keV]
Background photons
3000
Compton scattered photons from Fe 200
75
X-rays from Pb (K1)
25
X-rays from Sn (K1)

Absorption
material
Fe
Pb
Sn
Cu

Linear absorption
coefficient [cm1]
0.315
10.7
25
180

1 % thickness
[cm]
14.6
0.43
0.18
0.025

Shielded room
The most effective, convenient arrangement is a wholly shielded enclosure, to accommodate both the subject
and the detector system. For a given thickness of the chosen shielding material, this design offers the greatest
reduction in background and it offers also the smallest dependence of background response on body size.
Landolt-Brnstein
New Series VIII/4

10-64

10 Measuring techniques

[Ref. p. 10-81

Occasionally, subjects may react to isolation in a shielded room; such instances are much rarer where access
to the counter is via an open shielded labyrinth rather than through massive hinged or sliding doors. In
practice the background reduction factors are smaller than expected from the theoretical consideration
above, mainly due to scattering of high energy photons (E >3000 keV) down into the energy range of in
vivo counting (Table 10.17).
Table 10.17. Shielding parameters for some selected shielded rooms for in vivo measurement devices
Shielding materials

Reference
Background
Background
reduction factor index [cpm/cm3]
0.43
85Sch
70 cm silica sand, 0.3 cm Pb, 0.04 cm Cd, 0.1 cm Cu, 73
0.55 cm plastic
15 cm Fe, 1 cm Pb, 0.2 cm Fe
15 cm Fe, 0.9 cm Pb

100
40 - 57

0.36
0.34

85Sum
61Kie

Partial shielding
Some installations comprise more open structures, which eliminate direct paths for radiation between the
detector and the laboratory. Examples are the shadow shield design used for assessing whole-body
radioactivity in which the subject lies on a bed moving under a fixed detector in a central turret. Other
arrangements embodying the same principle can be devised to assess the radioactivity of individual organs or
regions. In another simple arrangement the detector and the back of a chair or bed holding the subject are
shielded for the investigation of radioactive deposits in larger regions. With all partially shielded counters the
background response below 200 keV is likely to be much larger than in a shielded room, because they
respond to photons scattered by the subject into the detector. For this reason a shielded room is essential for
the sensitive assessment of low-energy photon emitters. More information about shielding is given for
example in the IAEA Directory of whole-body radioactive monitoring [70IAE].
In addition to the shielding of the environmental radiation it is very important that all materials in the
detector systems, the mounting facilities and the shielding are selected for low level of intrinsic
radioactivity. Also the natural radioactivity in the construction materials of the surrounding building is
important for the background. The natural activity concentration of 40K can vary in between 200 Bq/kg
and 800 Bq/kg in bricks and 320 Bq/kg and 800 Bq/kg in cement, respectively [92Zik]. Thus, there is a
large potential for background reduction by proper selection of the construction materials. Moreover it is
important to minimise the amount of material close to the detector system in order to minimise the
background component due to Compton scattering in the direct vicinity of the detectors. Last but not least
there is a need for air filtration in order to reduce the background component due to airborne radioactive
materials.
10.3.2.4.3.2 Active methods
There is different kind of active methods for the reduction of the detector background signal, most of
them being based on anticoincidence techniques. Firstly this technique was applied for proportional
counters in order to reduce the background due to particles or Compton electrons from the environment.
For this purpose the counting volume of the proportional counter was surrounded by guard counters and
all coincident absorption events in both the counting volume and one of the guard counters were
discriminated. Thus only the events due to photoelectric absorption in the counting volume were
processed, this resulting in very good detection features especially for low energy photons such as the
plutonium L X-rays. On the other hand the sensitivity of these detectors for photons with higher energy as
for example the 59.6 keV -rays of 241Am is very low, even when the proportional counters were operated
with heavy counting gases such as Xe under high pressure of 2 or 3 bar. However, for in vivo
measurement of plutonium via the L X-rays it is essential to measure simultaneously the 241Am activity
present, because the L X-ray yield of 241Am is one order of magnitude higher than that of plutonium. The
energy resolution of the proportional counters does not allow for discrimination of the 241Am L X-rays
Landolt-Brnstein
New Series VIII/4

Ref. p. 10-81]

10 Measuring techniques

10-65

from the plutonium L X-rays and so the contribution from 241Am must be determined by measurement of
241
Am via the 59.6 keV -rays in the same measuring geometry. It was mainly due to this reason that the
proportional counters were replaced in most laboratories by the phoswich detectors, which were
commercially available since the late sixties.

Photo multiplier
Other source
of radiation

CsI (Tl)

(4)
NaI (Tl)
(1)

(2)

(3)

Be. entrance window


Source
material

Fig. 10.39. Measuring principle of a


phoswich detector.

The phoswich detector has been developed by Laurer for the in vivo measurement of low energy
photon emitters such as 210Pb, 239Pu and 241Am [68Lau]. The detector consists of a large area NaI(Tl)
crystal the thickness of which being just enough to fully absorb the low energy photons (typically
1-3 mm). The NaI(Tl) crystal is backed by a CsI(Tl) crystal for the detection of scattered photons due to
Compton effects in the NaI(Tl) crystal (Fig. 10.39). The photomultiplier tubes detect the scintillation light
from both crystals. However, because of the different scintillation decay times of the materials, it is
possible to assess by pulse shaping techniques whether the scintillation light is originating from the
NaI(Tl) crystal, the CsI(Tl) crystal or from both crystals. Thus it is possible to discriminate the Compton
scattering events (in both crystals) from the full absorption events, this resulting in a significant reduction
of the detector specific Compton continuum of the NaI(Tl) crystal. When comparing the count rates of a
phoswich detector in the low energy range (10 - 100 keV) with and without applying the pulse shape
discrimination technique a reduction of about one order of magnitude is observed. This figure, however,
does not correspond to the actual reduction of the Compton continuum from the NaI(Tl) crystal because
most of the scintillations detected by the photomultipliers are due to absorption events in the CsI(Tl)
crystal only. Actually the reduction of the NaI(Tl) Compton continuum is less than a factor 2 because the
CsI(Tl) anti Compton shield covers less than a 2 space angle.
Phoswich detectors provide good detection features for low energy photon emitters if no high-energy
photon emitters such as 137Cs are present. Compton continuum, backscatter peaks and characteristic X-ray
peaks due to those high-energetic emitters may influence the spectrum in the low energy region significantly,
this giving rise for problems in background prediction.

Landolt-Brnstein
New Series VIII/4

10-66

10 Measuring techniques

[Ref. p. 10-81

10.3.2.5 Spectrum evaluation


The interpretation of a photon-energy spectrum of body radioactivity will involve initially the identification of
radionuclides responsible for its individual features. The next stage, unless the spectrum is dominated by the
contributions from a single radionuclide, will generally involve resolution into the constituent components. In
a further process, the response attributable to a particular contributor will be translated into an estimate of
body or organ radioactivity; this is accomplished by reference to a spectrum representing a known
radioactivity of the nuclide measured in the same conditions.
Methods of deconvoluting photon-energy spectra of body radioactivity do not differ in principle from
those applied in X- and gamma-ray spectrometry generally, except that account must often be taken of the
effect on spectral shape of scatter in a large attenuating mass. The process is at its simplest in the estimation of
peak areas from semiconductor detectors. The good energy resolution of such instruments allows the effective
background response underlying a spectral peak to be reliably deduced from the adjacent continuum.
In the case of scintillation counters, the width of spectral peaks makes this approach often inapplicable
especially in case of multiple peaks. It will generally be necessary to first subtract an appropriate spectrum of
counter background and a more rigorous analytical procedure will generally be required.
The activity of the given radionuclide (q) can be expressed as follows
q=

where

N
t y

(10.3.2.3)

N is the number of net counts in the full energy peak area,


t is the measuring time,
y is the yield of (or X)- ray,
is the counting efficiency at the given energy for the respective measuring geometry

This is the most dominating evaluation method in gamma spectrometry.


Stripping method
When deconvolution of spectra from scintillation counters is required, a stripping process is sometimes
followed. Reference spectra are derived for each nuclide present, each representing the response from known
amounts of the nuclide in appropriate measuring geometry and in relevant absorbing media. The reference
spectrum containing the peak with the highest energy is selected, and it is normalised to the subject's spectrum
on the basis of count rate in an energy region where only that nuclide contributes. Subtraction of the
normalised spectrum gives a residue representing the remaining components, which is treated in the same
way. The activity of each radionuclide is calculated directly from the fraction of its reference spectrum, which
must be subtracted. In principle, the process can be repeated until the residue consists of the response from a
single nuclide only. In practice, unacceptable errors are likely to accumulate if the number of stages exceeds
two or three, particularly in relation to minor components in a spectrum; moreover, the method will generally
be inapplicable when the dominant peaks of different components overlap.
Linear regression analysis
A more satisfactory procedure in many situations is to adopt a method of linear regression analysis, to derive
the proportions of each reference spectrum which, when combined, gives rise to the best fit to the subject's
spectrum. Facilities for such analyses are embodied in several commercially available computer programmes
for processing ray spectra; alternatively, they can be developed locally. Utilizing a much larger portion of
the spectrum, instead of the restricted regions successively considered in the stripping process, this method
gives improved statistical accuracy in the estimates of the various components; moreover, realistic estimates
of this accuracy may be derived in the matrix-inversion procedures.
As with other methods of deconvolution, this approach has its limitations. In particular, it demands
stability of the spectrometer during the measurement, especially if the nuclides present possess overlapping
spectral features. It is also important that the locations of peaks in the subject's spectrum should coincide with
those in the relevant reference spectra; where minor drifts occur between the measurements, adjustments can

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-81]

10 Measuring techniques

10-67

often be made prior to the analysis if appropriate routines are available. However, the validity of the analysis
depends also on the spectral shapes of the reference standards according with those of the corresponding
components in the subject's spectrum.
This method is used practically exclusively in scintillation gamma spectrometry, however the procedure
can be extended to the deconvolution of any kind of complex distribution of measured data. Such application
is unfolding profile scanning data to quantify the measured distribution pattern in terms of activity deposited
in different body regions or organs.
10.3.2.6 Measuring geometries
In principle, the in vivo measuring systems can be allocated to two different types of systems, namely
geometry dependent and geometry independent systems. Here the geometry is defined as the detector
configuration in relation to the photon-emitting source in the body.
10.3.2.6.1 Geometry depending systems
Static geometry
The most common kind of geometry dependent systems are those having detectors that are positioned close to
the subject looking to specific organs or tissues. The advantages include high efficiency, better subject
positioning and less space requirement for the system. The use of such a static geometry dependent system is
extremely important in measuring low energy photons where the efficiency of the detection system needs to
be maximized.
Isotopes of iodine, and also 99mTc may concentrate in the thyroid gland. The range of photon energies
encountered is 27 keV (125I) to several hundred keV. Some HPGe detectors used for assessment of actinides
in lungs are of suitable diameter (ca. 50 mm) in relation to the size of the thyroid, and are large enough to
provide adequate detection efficiency over most or all of this energy range. Alternatively, a planar germanium
detector or thin NaI(Tl) crystal may be adopted for photon energies <100 keV, with thicker crystals, either
NaI(Tl) or co-axial germanium detectors, used if necessary to secure efficient photon detection at high
energies. More accurate assessment of an easily-detectable deposit would require better shielding, with the
detector recessed in a suitable collimator.
Inhalation is the most common route for intake in occupationally exposed personnel, with the respiratory
tract being the site of initial deposition. If the deposit persists for a sufficient time, monitoring of pulmonary
activity may offer the most sensitive and reliable means of assessing the intake. Indeed, in the case of certain
actinides which subsequently re-locate to organs absorbing virtually all low-energy photon emissions, it offers
the only remotely practicable means of assessing an intake by external counting. A large-diameter (150-300
mm) stationary NaI(Tl) detector recessed in a cylindrical collimator may be used. The response would
preferably be recorded in two locations, the detector viewing in turn the anterior and posterior surfaces of the
thorax; ideally, if two such detectors were available, the measurements could proceed simultaneously. The
sensitive measurement of pulmonary deposits of low-energy photon emitters (<100 keV) is most commonly
required for 241Am and for isotopes of uranium and plutonium. It requires equipment giving a better signal-tobackground ratio than is provided by NaI(Tl) detectors. This is achieved either through partial suppression of
background response as in the phoswich detector, or through the improved energy resolution offered by
semiconductor detectors [00Lop]. Fig. 10.40 - Fig. 10.42 show as typical examples the arrangements of 2
phoswich detectors and 4 HPGe detectors as used in the Research Centre Karlsruhe for the in vivo
measurement of low energy photon emitters in the lungs.

Landolt-Brnstein
New Series VIII/4

10-68

10 Measuring techniques

[Ref. p. 10-81

Fig. 10.40. Typical arrangement of 2 phoswich detectors (Harshaw 20 cm diam. 1 mm NaI(Tl) / 50 mm CsI(Tl)
crystals) for in vivo measurement of low energy photon emitters in the lungs

Two other sites of deposition, which frequently attract specific interest, are bone and liver. Arrays of
detectors viewing the skull have been employed to assess skeletal deposits of 210Pb (47 keV photons) and
241
Am (59.6 keV). The levels of contamination in liver and skeleton may also be of interest in regard to their
interfering contributions when lung deposits are assessed.
An extreme, but fairly common example of a localized deposit is the presence of poorly soluble
radioactive material at the site of a puncture wound, investigated shortly after an accident, before important
quantities have become systemic. With fission or activation products giving abundant and energetic photon
emissions and with high limits of intake, improvised arrangements employing any spectrometrically suitable
scintillation or semiconductor detector are likely to be satisfactory.
Profile scanning geometry
Another kind of geometry dependent systems is using moving detectors or subjects. The so-called scanning
systems can be used for identifying the organ or tissue where the radionuclide in question is deposited. The
so-called profile scanning systems are providing information on the activity distribution pattern in the body.
Better spatial resolution can be obtained by using collimators in the front of the detectors however it can only
be done at the expense of the counting efficiency. Profile scanning measurement may also point to the
presence of surface contamination, which has to be removed.
It should be noted that profile scanning with simple slit-collimated detectors would often give reliable
indications of the relevant sites of deposition, but that only a rough evaluation of that deposition can be
obtained if a single detector viewing only one aspect of the body is employed. Quantitative assessment of
radionuclide distribution by profile scanning requires either paired detectors or, if only one is available,
separate traverses of the anterior and posterior surfaces, and further improvements may result if focused
collimators are used and computer-aided evaluation techniques are applied.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-81]

10 Measuring techniques

10-69

Fig. 10.41. Typical arrangement


of 4 HPGe semiconductor
detectors (Silena HPGe sandwich
with 20 cm diam. planar p-type
HPGe crystal backed by 50 %
coaxial n-type HPGe crystal) for
in vivo measurement of low
energy photon emitters in the
lungs.

10.3.2.6.2 Geometry independent systems


Geometry independent systems are characterized by the detectors being located in a certain distance from the
subject to be measured, either above or below or both. In those arrangements the detectors have an almost
uniform response for any distribution of gamma emitters in the body. Thus, arrangements characterized by
this geometry independent feature were originally called as whole-body counter.
Arc geometry
This arrangement is capable of high accuracy if the levels of internal radioactivity and the space available
inside a shielded room are sufficient. The subject lies on a curved frame forming the arc of a circle centered at
the detector, so that all parts of the body are roughly equidistant from it. The detection efficiency is of course
poor and, even with a large detector in a heavily shielded room, the technique will not generally be applicable
to the determination of body burdens below several kBq; it would seldom be feasible with semiconductor
detectors. In the context of radiological protection, therefore, the arc method will be used primarily in the
investigation of established cases of internal contamination rather than as a regular means of control.

Landolt-Brnstein
New Series VIII/4

10-70

10 Measuring techniques

[Ref. p. 10-81

Chair geometry
In this arrangement the subject reclines in a tilted chair with the detector supported typically 0.4 m above the
abdomen. The detector will commonly be a NaI(Tl) scintillator, e.g. 200 mm diameter 100 mm, and in a
heavily shielded room would be capable of detecting as little as 50 Bq of most common fission and activation
products in a counting time of 15 min. Similar arrangements have been employed with semiconductor
detectors. The response will however depend markedly on the location of the radioactive deposit, e.g. in the
lungs or liver, may differ by a factor of two or more from that applying to material widely dispersed in the
body. The potential for systematic error is accordingly much greater than with the arc technique, but this will
be unimportant in many routine applications. Recently, fully automated chair type whole body counters have
been developed providing very good detection features [02Sin]. The chair geometry is also applied in most
of the mobile whole body counters used for monitoring of staff of nuclear power plants.
Scanning geometry
With this design of counter, the response is accumulated from a single detector while traversing the subject's
length at a fixed distance above or below the supine body, or in some versions, in a corresponding disposition
relative to the erect body. Alternatively, the supine subject may be moved in relation to a fixed detector.
Accuracy will be improved if a second traverse is performed with the subject's posture reversed and the
evaluation is based on the combined response in the two positions. When employing two detectors (one above
and one below the supine subject), a more representative sample of the photon flux is obtained by moving the
supine subject relative to the counters during accumulation of the spectrum.
However, while scanning arrangements can provide more uniform detection geometry compared with that
given by most other configurations, they might not yield significantly greater accuracy than a well-designed
multi detector array of stationary detectors. Useful indications of the distribution of a radionuclide within the
body may be secured if the system can display a profile of the response according to position.
Stretcher geometry
An alternative approach, preserving the good detection efficiency given by the chair technique but offering
improved uniformity of response, is to adopt a stretcher geometry, with the subject in a supine posture and
several detectors distributed about the body. Commonly 4-8 detectors are employed, disposed above and
below the stretcher so that their combined response is acceptably independent of the source distribution.
Complete uniformity of efficiency is of course unattainable, however, for activity, which is not concentrated
in small organs or regions, such an array can yield results for energetic (>100 keV) photon emitters accurate
to within 20 % or better. Fig. 10.42 shows as an example a stretcher type whole body counter with 4 NaI(Tl)
scintillation detectors as used in the Research Centre Karlsruhe for in vivo measurement of photon emitters
with energy between 100 and 3000 keV.
Disadvantages are the need to provide several independently adjustable supporting mechanisms for the
various detectors, and the requirement of a larger shielded room than would generally be necessary to
accommodate only a chair.
Table 10.18. Comparison of various whole-body counter geometries
Geometry

Mechanical
arrangement

Uniformity
of response

Information on Sensitivity
distribution

MDA of 137Cs.
[Bq]

Arc

fixed

very good

no

low

300

Chair

fixed

poor

no

high

100

Static array

fixed

good

possible

high

70

Scanning
(collimated
detectors)

moving detector or
moving bed

good

yes

high

100

Shadow shield

moving bed

good

yes

medium

130

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-81]

10 Measuring techniques

10-71

Fig. 10.42. Stretcher type whole body counter with 4 NaI(Tl) scintillation detectors (Bicron 20 cm diam. 10 mm
crystals) for in vivo measurement of medium energy photon emitters (100 - 3000 keV)

10.3.2.7 Calibration
The purpose of whole-body counter calibration is to determine the relationship between the detector
response and the radioactivity in the body. Gamma emitting radionuclides can be characterised by the
shape of their spectra or by the location of spectral peaks, which correspond to emitted photon energies.
In certain circumstances, for example when spectra from scintillation counters are analyzed by leastsquares fitting, it is essential to use the same nuclides for calibration as those to be measured. In the
situations it may suffice to derive a calibration factor by interpolation of data measured individually for a
series of monoenergetic photon emitters covering the energy regions of interest. With high-resolution
semiconductor detectors, a single nuclide emitting photons at several energies, or a suitable mixture of
nuclides, may be more convenient.
10.3.2.7.1 Energy calibration
As has already been mentioned energy calibration establishes the relationship between spectral peak
location (channel number) and emitted gamma ray energy. This relationship may then be used to
identify radionuclides from the location of their spectral peaks. Energy calibration is performed using
radioactive sources emitting gamma rays with known energies. The gamma energies emitted by the
calibration source(s) should cover the range of energies likely to be encountered during whole-body
measurements.

Landolt-Brnstein
New Series VIII/4

10-72

10 Measuring techniques

[Ref. p. 10-81

Table 10.19 lists a selection of radionuclides suitable for calibration purposes. The analysis systems in
modern whole-body counters automatically derive the energy calibration relationship using fitting
routines and also plot the function for inspection.
Many software techniques require that the peak shape has to be characterised as a function of
energy. Even if the software does not require it, peak shape is a useful tool both to confirm proper
operation of the detector and to identify the presence of multiple nuclides with similar gamma
energies. Peak shape calibration, characterised by FWHM (Full width at half maximum height), is
usually performed at the same time and with the same spectrum as energy calibration.
Table 10.19. Characteristic parameters of radionuclides suitable for photon energy and efficiency
calibration
Nuclide Half-life

Photon emission
Energy
[keV]

55

Fe

241

109

57

Am

Cd

Co

141

Ce

Nuclide Half-life

5.89 (Mn K2)


5.90 (Mn K1)
6.49 (Mn K1)

Emission
probability
[%]
8.4
16.6
3.4

13.93 (Np L)
17.61 (Np L)
21.00 (Np L)
26.35
59.54

13.2
19.4
4.9
2.4
36.0

463 d

88.03

3.65

271.84 d

122.06
136.47

85.59
10.58

145.44

48.9

2.73 y

432.0 y

32.50 d

Energy
[keV]

137

Ce

137.65 d

165.85

80.0

203

Hg

46.61 d

279.20

81.3

51

Cr

27.71 d

320.08

9.58

22

Na

950.4 d

511.00

180.7

1274.54

Emission
probability
[%]
99.94

Cs

30.0 y

661.66

85.0

54

Mn

312.5 d

834.84

99.98

46

Sc

83.80 d

889.28
1120.55

99.98
99.99

60

Co

1925.5 d

1173.24
1332.50

99.90
99.98

88

106.66 d

898.04
1836.06

94.6
99.24

4939 d

121.78
244.69
344.37
778.89
963.38
1085.78
1112.02
1407.95

28.37
7.51
26.58
12.96
14.62
10.16
13.56
20.58

152
139

Photon emission

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-81]

10 Measuring techniques

10-73

10.3.2.7.2 Efficiency calibration


Efficiency calibration allows one to convert the measured count rate in a region of the spectrum to the
emission rate of the corresponding gamma photon of the radionuclide. Emission rate can then be
converted to disintegration rate or activity using the probability of photon emission per decay. The
conversion factor between count rate and activity is energy dependent and must be measured for the range
of photon energies, which are expected to be present in the whole-body measurements. The variation in
the efficiency factor as a function of photon energy may be obtained by simple interpolation between the
measured points on the spectrum or by fitting an empirical function to the points. If one knows in advance
the radionuclide to be measured, a situation frequently encountered in medical applications, then the
efficiency factor is simply determined by using the same nuclide for calibration. In general, however,
more complex spectral techniques are needed to derive efficiency factors for the spectra encountered in
radiation protection applications. Different spectral analysis techniques should be applied to high or lowresolution detector systems.
To achieve accuracy in calibration the following factors must be observed:
The same detector and hardware configuration should be used both for calibration and whole-body
counting; the possible influence of necessary changes on the detection efficiency must be checked.
Identical spectral analysis methods must be used.
The source - detector geometry must be adequately simulated.
The photon attenuation conditions applying in the body should be adequately reproduced
As a general rule the lower the energy of the photons to be detected the greater the care required to
achieve a specified accuracy.
10.3.2.7.2.1 Point source calibration
This procedure may be adopted for whole-body counters whose geometrical counting efficiency shows
little dependence on the location of the source, for example those disposed in distant arc geometry and in
certain scanning arrangements. With the arc geometry, calibration accurate to within a few percent can be
achieved with a standardised point source suspended in a tank of water or located between stacked plates of a
solid absorber with comparable attenuation properties, the appropriate location of the source in the tank or
stack is deduced from examination of the spectral shape and evaluation of the relative photon fluxes emerging
from the subject's surfaces.
10.3.2.7.2.2 Phantom calibration
In practice systemic radionuclides tend not to be concentrated in a single anatomical region and such counters
are generally calibrated, at least in the first instance, with the aid of whole-body phantoms simulating the
human form and containing a standardised aqueous solution of the relevant nuclide. The most convenient
general-purpose phantom is the so-called BOMAB (BOdy Manikin ABsorption) phantom, which consists of
a collection of polyethylene vessels of circular or elliptical cross-section (Fig. 10.43). These are available
commercially, or can be made by a workshop with experience of plastics. It is useful to have available two or
three such phantoms with dimensions suitably scaled to represent individuals of different sizes; for
intermediate physiques calibration factors may be derived by interpolation according to functions of
anatomical parameters e.g. weight/height.

Landolt-Brnstein
New Series VIII/4

10-74

10 Measuring techniques

[Ref. p. 10-81

Fig. 10.43. The BOMAB phantom for simulation of homogeneous activity depositions in the whole body
1.5
1.4

Counting efficiency [%]

1.3
1.2
1.1
1.0
0.9
0.8

Fig. 10.44. Counting efficiency of a standard whole body


counter (stretcher type with 4 NaI(Tl) scintillation detectors
as shown in Fig. 10.43) for homogeneous activity
depositions in the whole body according to calibration
with the BOMAB phantom.

0.7
0.6
0.5
0

500

1000
1500
Photon energy [keV ]

2000

Alternatively makeshift arrangements, e.g. using plastic reagent bottles, may suffice; and in some
situations more versatile in simulating specific physiques and postures (Bottle phantom). Conversely, much
more elaborate whole-body phantoms (REMCAL, REMAB) can be purchased, some of them provided with
discrete organs, which can be labelled independently of a dispersed deposit. Such complicated devices would
ordinarily be required only in the calibration of equipment for assessment of deposits in specific organs.
Recently, phantoms were developed which do need to be filled by water solution of radionuclides.
Organic gels with dissolved radionuclides are used for the filling of BOMAB phantoms or large number
of point sources is inserted into polyethylene bricks from which phantoms of different body height and
weight could be easily built (IGOR). Comparison of various phantom characteristics is shown in
Table 10.20

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-81]

10 Measuring techniques

10-75

Table 10.20. Comparison of the characteristics of different phantoms used in whole-body counting.
Type of
Phantom

Availability

Price

Versatility Antropomorphity

Handling

Decontamination

Bottles

very good

cheap

good

satisfactory easy

not necessary

BOMAB

good

medium

bad

satisfactory easy

difficult

REMCAL

good

very expensive

good

good

complicated

very difficult

REMAB

good

very expensive

good

good

complicated

very difficult

Presswood

very good

cheap

bad

bad

easy

not necessary

Besides phantoms for whole-body counting there are several phantom constructions simulating certain
body regions such as thyroid in neck phantom (IAEA/ANSI neck phantom) or chest phantom containing
organs like lungs, pulmonary lymph nodes, liver etc. (LLNL realistic chest phantom, JAERI phantom for
Asian men). These latter phantoms were very sophisticatedly constructed which are especially suitable for
calibration of low energy photon emitting radionuclides like transuranium elements deposited in the
human respiratory tract. The LLNL chest phantom has been developed at the Lawrence Livermore
National Laboratory and then manufactured by Humanoid Systems Inc. (Fig. 10.45). The phantom made
from tissue equivalent materials represents the thorax of a male adult with 177 cm height and 76 kg
weight. Lungs, tracheo-bronchial lymph nodes and the liver can be exchanged by active components with
homogeneous deposition of plutonium and other low energy photon emitters. Overlay structures are also
provided for the simulation of chest walls with different thickness (1.6 - 4.1 cm) and different
muscle/adipose composition. At present there are 3 generations of the LLNL phantom, which are used as
international standard for lung counter calibration [02Kra]. However, the LLNL phantom cannot be used
for calibration for inhomogeneous deposition in the lungs or other organs. For this reason a second
anthropomorpheous phantom has been provided by Humanoid Systems with the organs having a hole
matrix for inserting small cylindrical sources in order to simulate any kind of inhomogeneous organ
deposition (Fig. 10.46).

Fig. 10.45. The LLNL chest phantom for simulation of homogeneous activity depositions in the lungs, tracheobronchial lymph nodes and in the liver (Humanoid Systems Inc.)

Landolt-Brnstein
New Series VIII/4

10-76

10 Measuring techniques

[Ref. p. 10-81

Fig. 10.46. The Fission product phantom for simulation of activity depositions in all relevant organs of the trunk
(Humanoid Systems Inc.)

10.3.2.7.2.3 In vivo calibration


On occasion, known activities of short-lived tracers have been administered to volunteers expressly in order
to calibrate whole-body counters, e.g. 132Cs and 42K. It may sometimes be possible to make use of subjects
who have received intakes in medical diagnosis or other metabolic studies. This would only be possible under
supervision of ethics committee. If the measurement is delayed after intake of the tracer, e.g. to allow its
distribution to stabilise, excreta voided in the interim may need to be collected and assessed for their content
of radioactivity. When taking into account the loss of activity, the results of in vivo calibration are in very
good agreement with the results of phantom calibration, as demonstrated by Kaul for 42K [64 Kau].
10.3.2.7.2.4 Mathematical calibration
Methods of calibration using phantoms are relative methods; absolute methods do not require a
radioactive standard for calibration, however standards must be used to confirm a calibration.
Mathematical phantoms, using Monte Carlo method may be used for such calibration, as demonstrated by
Mallet [95Mal]. The advantage of such phantoms is that any distribution of the radionuclide in the
phantom could be used. However, thorough comparison of calculated examples with the measured values
has to be performed as to ensure good quality of the mathematical phantom. It is especially important
when low energy gamma emitters are subject of interest and the radionuclide in the body is nonhomogeneously distributed, as shown by Hunt for some cases with 241Am in the lungs and at the bone
surface or with natural uranium in the lungs, respectively [03Hun].

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-81]

10 Measuring techniques

10-77

Fig. 10.47. The Yale phantom as used by Hunt for the Monte Carlo simulation of the radiation transport from the
activity deposition in the body (i.e. 241Am at the bone surface) to the detector outside the body (i.e. 8 4 NaI(Tl)
scintillation detector over the lungs) [03Hun].

10.3.2.8 Uncertainties and detection limits


Generally, the uncertainties in the measurement are difficult to estimate. When activity levels are low and
close to the limit of detection, uncertainties due to counting statistics may dominate the overall
uncertainty. For radionuclides that are easily detected and present in sufficient quantity, uncertainties due
to counting statistics will be small compared to other sources of uncertainty. Consideration must also be
given to systematic uncertainties in other parts of the measurement procedure, e.g. calibration, or
correction for body size of in vivo measurements, etc. Typically, the components of uncertainty are
grouped in two categories: Type A comprises those components, which can be described by the Poisson
distribution (i.e. counting error, to some extend also the variation of background signal and the variation
of the subject positioning) whereas Type B comprises all other components (i.e. variation of body
dimensions, overlaying structures, distribution of activity within the body, and the uncertainty of the
calibration standard). The Type B components cannot be expressed in terms of Poisson statistics, and thus
they cannot be associated with the Type A components in order to derive the total uncertainty of the
measurement. Table 10.21 lists some typical values for the various components of uncertainty.
Table 10.21. Typical values for the components of uncertainty for the in vivo measurements of
radionuclides emitting low, intermediate and high photon energy radiation
Uncertainty (%)
Source of uncertainty (Type)
Low photon energy Intermediate photon High photon energy
E <20 keV
energy
E >100 keV
20 keV < E <100 keV
Counting statistics (A)
50 %
30 %
7%
Variation of detector positioning (A) 20 %
5%
<5 %
Variation of subject background (A) 50 %
10 %
<5 %
Variation in body dimensions (B)
50 %
12 %
7%
Variation of overlaying structures (B) 30 %
15 %
12 %
Variation of activity distribution (B) 30 %
5%
<5 %
Calibration (B)
5%
5%
5%
Spectrum evaluation1) (B)
15 %
5%
3%
1) HPGe detector spectra

The statistical quantities for describing the Type A uncertainty and the corresponding detection limits
are analogous to those used in all other kind of radioactivity measurement. Therefore, in recent years
mainly the ISO definitions [98ISO] are applied for the calculation of uncertainties and detection limits of
in vivo measurements. There are two basic terms being complementary to each other, i.e. decision
threshold and detection limit.

Landolt-Brnstein
New Series VIII/4

10-78

10 Measuring techniques

[Ref. p. 10-81

Decision threshold
The decision threshold (frequently also referred to as decision level or critical level) is an a posteriori
calculated value at which the decision can be made, whether the registered pulses include contributions
from the body or are solely due to background. If this decision rule is observed, a wrong decision that
there is a contribution from the body when actually only a background effect exists (Type I error), occurs
with a well-defined probability . By definition the decision threshold is given in terms of pulses but for
practical application it is frequently transferred to the corresponding activity value.
Detection limit
The detection limit (frequently also referred to as minimum detectable activity or lower limit of detection)
is an a priori calculated value, which specifies the minimum body contribution that can be detected by a
defined measurement procedure. The detection limit is complementary to the decision threshold, i.e. when
considering the detection limit the wrong decision that there exists only a background effect when there is
in fact a contribution from the body (Type II error), occurs with a well-defined probability . Thus, the
detection limit is closely related to the decision threshold defined by the Type I error probability . By
definition the detection limit is given in terms of body or organ activity and it can be compared directly
with guideline values.
The choice of the values of and depends on the aim of the measurement. For the purpose of
radiation protection typically the values = = 0.05 are used (i.e. 5% probability for both Type I and
Type II errors). With these values the following formula for the decision threshold may be derived from
the general concept given by Altshuler and Pasternack [63Alt]:
N DT = 1.645 N B

where
NDT
NB
tB
tS

tS
tB

tS
1 +
t
B

(10.3.2.4)

is the decision threshold in terms of net counts in the full energy peak region for = = 0.05,
is the total number of background counts in the full energy peak region,
is the background measuring time,
is the subject measuring time,

For the detection limit the generic formula derived by Currie [68Cur] may be used:
N DL = 2.71 + 2 N DT

(10.3.2.5)

where
NDL is the detection limit in terms of net counts in the full energy peak region for = = 0.05.
When the background count rate is high enough, Eq. (10.3.2.5) can be simplified and the following
expression is derived for the detection limit:
q DL =

t
3.3
NB S
tB
tS y

tS
1 +
tB

(10.3.2.6)

where
qDL is the detection limit in terms of body or organ activity for = = 0.05
y
is the yield of (or X)- ray,

is the counting efficiency at the given energy for the respective measuring geometry

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-81]

10 Measuring techniques

10-79

Table 10.22. Typical detection characteristics of a standard whole body counter (stretcher type with 4
NaI(Tl) scintillation detectors as shown in Fig. 10.40) for homogeneous depositions of some selected
fission and activation products in the whole body (subject counting time tS = 300 s; background counting
time tB = 1800 s)
Nuclide
57

Co
Cs
134
Cs
60
Co
22
Na
137

Detected photon radiation


Energy [keV] Yield
y [%]
123
86
662
85
796
85
1173
100
1275
100

Counting
efficiency
[%]
1.5
0.9
0.81
0.66
0.61

Background
count rate
NB/tB [cps]
14.0
7.4
4.9
3.9
4.0

Lower limit of detection [Bq]


Required
Achieved
(Table 10.12)
20000
60
10000
73
6000
66
1000
62
200
67

Table 10.23. Typical detection characteristics of a lung counter (4 HPGe semiconductor detectors as
shown in Fig. 10.3.2.7) for homogeneous depositions of some selected actinides in the lungs (HPGe AntiCompton counter of the Nuclear Research Centre Karlsruhe; subject counting time tS = 3000 s;
background counting time tB = 30000 s)
Detected photon radiation Counting
Lower limit of detection [Bq]
Nuclide
Background
count rate
Energy [keV] Emission pro- efficiency
Required
Achieved
[%]
NB/tB [cps]
bability [%]
(Table 10.12)
239
Pu
2.3
0.017
0.0088
2
1500
17.1(L)
241
Am
59.6
35.9
0.47
0.0090
0.2
3.6
235
U
186
57
0.24
0.0041
3
3.0
1) Calibration with LLNL chest phantom for 25 mm chest wall thickness and 50/50 muscle/adipose tissue composition

10.3.2.9 Measurement procedure


Subjects for direct measurements should be free of external surface contamination and in fresh clothing,
often disposable paper garments. Accessories such as jewellery, watches and spectacles should be
removed. Such precautions help to avoid false identifications of internal activity, and also prevent the
transfer of contamination to the counting equipment. Individuals should, to the extent practicable, be in a
defined counting position, to ensure reproducibility in serial measurements and to improve comparison
with calibration results. In some cases the subject will need to remain stationary for periods up to an hour
for satisfactory precision in the measurement. Some means of communication should be provided for
subjects in enclosed shielding, particularly when extended counting periods are necessary.
For counting systems based on scintillation counting (NaI(Tl) crystals or phoswich detectors),
background counts for the detector system should therefore be determined using an appropriate phantom,
as similar as possible to the subject to be counted and placed in the defined counting position. For whole
body counting, background counts determined using uncontaminated subjects matched with respect to
gender, height and weight would improve results. Measurements of background in the counter should be
made as close as possible in time to the measurement of the subject, ideally just before and just after.
When using semiconductor detectors, background counting with matching phantoms is not necessary.
10.3.2.10 Quality assurance and control
Quality assurance (QA) for the measurement of internally deposited radionuclides includes all steps
necessary to confirm the accuracy of the measurement and the validity of the dosimetrc interpretation.
Guidance for quality assurance is provided by international institutions such as the International
Standards Organization as well as by national authorities. The nature and extent of the QA programme
should be consistent with the number of workers monitored, and the magnitude and likelihood of
exposures expected in the workplaces to be covered by the monitoring programme.
Landolt-Brnstein
New Series VIII/4

10-80

10 Measuring techniques

[Ref. p. 10-81

All persons involved in the internal exposure assessment programme are responsible for its quality
and therefore for implementing its QA programme and quality control (QC) procedures. Responsibility
for the quality of a particular operation should be delegated to the person actually performing the
operation. Such persons should be actively involved in the development of QC procedures, and trained in
methods of detecting non-compliance.
A direct measurement facility should have a designated QA representative. This representative should
monitor QC procedures, perform internal audits of the programme, and be responsible for training all
personnel in QA, both in general terms and in the specific quality aspects of their individual work.
The fundamental requirements for a complete programme of QA include:

compliance with general operational requirements stated in accepted written criteria


a clearly documented in-house QA program
periodic performance evaluations, including proficiency measurement tests
documented procedures and quality assurance programme for services provided to customers

Quality control
Quality control in a measurement process is important to ensure that assessment of intakes is as reliable as
possible. Evidence of the validity of such assessments may be required for legal and/or regulatory purposes.
Quality control programmes include the following activities:

procedures and protocols for proper management of the internal dosimetry programme,
detector system verification,
routine verification of proper instrument performance,
data recording and archiving,
audits and accreditation,
intercomparison [01Ram, 00Doe].

Procedures and protocols


The laboratory should prepare and maintain an operational manual that outlines responsibilities and
provides requirements for data control, document control, maintenance/test, equipment calibration and
checks, procedure, training, corrective action in the event of non-compliance, and traceability to
standardizing bodies. The operations manual should include procedures to verify that the quality of the
measurements meets the appropriate accuracy requirements. The quality control procedures should be
carried out at appropriate intervals.
Performance checks
Performance checks of the system include energy calibration, energy resolution measurement and
determination of the relative counting efficiency, generally using a point source.
National regulations may require that facilities concerned with measurement and internal dose
assessment be accredited. Such accreditation programmes will have specifications for QA and QC to be
implemented.

Landolt-Brnstein
New Series VIII/4

10 Measuring techniques

10-81

10.3.2.11 References for 10.3.2


27Blu
29Sch
31Sch
37Eva
41Raj
51Sie
57And
57Sie
58Bir
61Kie
61Lan
62Bur
63Alt
64Kau
64Raj
68Cur
68Lau
70IAE
72Par
75And
75ICR
76Fal
76Sch
78New
79Gri
80Tai
83ICR
83Too
85And
85New
85Sch
85Sum
87ISO

Blumgart, H.I., Weiss, S.: J. Clin. Invest. 4 (1927) 389.


Schlundt, H., Barker, H.H., Flinn, F.B.: Am. J. Roentgenol. 21 (1929) 345.
Schlundt, H., Nerancy, J.T., Morris, J.P.: Am. J. Roentgenol. 26 (1931) 112.
Evans, R.: Am. J. Roentgenol. Radium Ther. 37 (1937) 368.
Rajewsky, B.: Strahlentherapie 69 (1941) 438.
Sievert, R.M.: Ark. Fys. 3 (1951) 337.
Anderson, E.C.: Br. J. Radiol., Suppl. 7 (1957) 27.
Sievert, R.M., Hultqvist, B.: Br. J. Radiol., Suppl. 7 (1957) 1.
Bird, P.M., Burch, P.R.J.: Phys. Med. Biol. 2 (1958) 217.
Kiefer, H., Maushart, R.: Kerntechnik 3 (1961) 228.
Langham, W.H., in: Radioactivity in Man. Thomas, G.C. (ed.), Springfield, IL: Charles C.
Thomas, 1961, p. 311.
Burch, P.R.J., Hughes, D., Hnuma, T.A., Overton, T.R., Appleby, D.B., in: Proc. IAEA Symp.
On Whole Body Counting, Vienna, Austria, 12-16 June 1961, Vienna: IAEA, 1962, p. 59.
Altshuler, B., Pasternack, B.: Health Phys. 9 (1963) 293.
Kaul, A., Schoeppe, W., Koch, K.M., Hierholzer, K.: Biophysik 2 (1964) 87.
Rajewsky, B., Kaul, A. and Heyder, J., In: Assessment of Radioactivity in Man 1, Vienna:
IAEA STI/PUB/84 (1964) 15.
Currie, L.A.: Anal. Chem. 40 (1968) 586.
Laurer, G.R.: The in vivo measurement of lung burdens of radionuclides emitting soft,
penetrating radiations, available from: UMI, 300 N. Zeeb Rd. Ann Arbor, MI, USA, Order
6904570, 1968.
International Atomic Energy Agency. Directory of whole-body radioactive monitoring.
STI/PUB/213, Vienna: IAEA, 1970, ISBN 02-0-112070-2.
Parr, R. M., Dudley, R. A., Fedorov, G. A., In: Assessment of Radioactive Contamination in
Man, Vienna: IAEA STI/PUB/290 (1972) 215.
Andrasi, A., Kotel, G.: Int. J. Appl. Radiat. Isot. 26 (1975) 451.
International Commission on Radiological Protection, ICRP Publication 23, Oxford and New
York: Pergamon Press, 1975.
Falk, R.B., Tyree, W.H., Wood, C.B., Lagerquist, C.R.A., in: Advances in Radiation
Protection Monitoring. Proceedings of a Symposium, Stockholm, Sweden, 26-30 June 1978,
Vienna; IAEA, 1979, p. 445.
Schmitt, A., Fessler, H., in: Diagnosis and Treatment of Incorporated Radionuclides.
Proceedings of a Symposium. STI/PUB/411, Vienna: IAEA, 1976, p. 285.
Newton, D., Fry, F.A., Taylor, B.T., Eagle, M.C., Sharma, R.C.: Health Phys. 35 (1978) 751.
Griffith, R.V., Dean, P.N., Anderson, A.L., Fisher, J.C., in: Advances in Radiation Protection
Monitoring, Vienna: IAEA, 1979, p. 493.
Tait, W.H.: Radiation Detection, London: Butterworth & Co., 1980.
International Commission on Radiological Protection, ICRP Publication 38. Ann. ICRP 11-12,
Oxford and New York: Pergamon Press, 1983.
Toohey, R.E., Keane, A.T., Rundo, J.: Health Phys. 44, Suppl. 1 (1983) 323.
Andrasi, A. Beleznay, E. and Urban, J. In: Assessment of Radioactive Contamination in Man,
Vienna: IAEA STI/PUB/674 (1985) 165.
Newton, D., Wells, A. C., Mizushita, S., Toohey, R. E., Sha, J. Y., Jones, R., Jefferies, S. J.,
Palmer, H. E., Riekst, G. A., Anderson, A. L., Campbell, G. W., In: Assessment of Radioactive
Contamination in Man, Vienna: IAEA STI/PUB/674 (1985) 183.
Shen, C., Wen, H., Zheng, W., Zhao, Y., Tang, M., Ye, C., Wu, D., In: Assessment of
Radioactive Contamination in Man. Proc. Symp. Paris, France, 19-23 November 1984, Vienna:
IAEA STI/PUB/674 (1985) 123.
Sumerling, T.J., McClure, D.R., Massey, D.K.: NRPB-R188, London: HMSO, 1985.
International Organization for Standardization, International Standard ISO9000, Geneva: ISO,
1987.

Landolt-Brnstein
New Series VIII/4

10-82

10 Measuring techniques

88ICR International Commission on Radiological Protection, ICRP Publication 54. Ann. ICRP 19 (13), Oxford and New York: Pergamon Press, 1988.
91Too Toohey, R.E.: Health Phys. 60, Suppl. 1 (1991) 7.
92Del Delaney, C.F.G., Finch, E.C.: Radiation detectors, Oxford: Clarendon Press, 1992.
92Zik Zikovski, L., Kennedy, G.: Health Phys. 63 (1992) 449.
94And Andrasi, A., Henrichs, K., Bogner, L.: Report EUR 15395 EN, 1994.
94IAE International Atomic Energy Agency. TECDOC-746, Vienna: IAEA, 1994.
94ICR International Commission on Radiological Protection, ICRP Publication 68. Ann. ICRP 24(4),
Oxford and New York: Pergamon Press, 1994.
94Skr Skrable, K. W., Chabot, G. E., French, C. S., LaBone, T. R.: Internal Radiation Dosimetry
(RAABE, O.G., Ed.), Medical Physics Publishing, Madison, WI (1994) 431.
95Doe Doerfel, H.: Proc. Symposium on Radiation Protection in Neighboring Countries in Central
Europe, Portoroz, 1995.
95Hub Hubbel, J.H., Seltzer, S.M.: National Institute of Standards and Technology, NISTIR 5632,
1995.
95Mal Mallett, M.W., Hickman, D.P., Knuchen, D.A., Poston, J.W.: Health Phys. 68 (6) (1995) 773.
96HPS Health Physics Society: Performance Criteria for Radiobioassay: American National Standards
Institute HPS N13.30-1996, McLean, VA: Health Physics Society, 1996.
96IAE International Atomic Energy Agency.: Safety Series No. 114, Vienna: IAEA, 1996.
96Kra Kramer, G.H., Loesch, R.M., Olsen, P.C.: Proc. 1996 International Congress on Radiation
Protection, Vienna,. 2 (1996) 409.
97ICR International Commission on Radiological Protection: ICRP Publication 78. Ann. ICRP
27(3/4), Oxford and New York: Pergamon Press, 1997.
98ISO International Organization for Standardization: International Standard ISO/WD 119298/ISO/TC85/SC2/WG5, Geneva: ISO, 1998.
99Kno Knoll, G.F.: Radiation Detection and Measurement, 3rd edn, New York: John Wiley & Sons,
1999.
99USD United States Department of Energy, DOE Standard DOE-STD-1112-98, 1999.
00And Andrasi, A.: Radiat. Prot. Dosim. 89 (3-4) (2000) 229.
00Doe Doerfel, H., Andrasi, A., Bailey, M.R., Birchall, A., Castellani, C.-M., Hurtgen, C., Jarvis, N.,
Johansson, L., LeGuen, B., Tarroni, G.: Forschungszentrum Karlsruhe GmbH, FZKA 6457,
2000.
00Lop Lpez Ponte, M.A., Bravo, T.N.: Radiat. Prot. Dosim. 89 (3-4) (2000) 221.
01Ish Ishigure, N., Nakano, T., Enomoto. H.: Radiat. Prot. Dosim. 97 (3) (2001) 271.
01Ram Ramzaev, V., Ishikawa, T. Hill, P., Rahola, T., Kaidanovsky, G., Yonehara. H., Hille, R.,
Uchiyama, M.: Radiat. Prot. Dosim. 98 (2) (2002) 179.
02Kra Kramer, G.H., Hauck, B.M.: Radiat. Prot. Dosim. 102 (4) (2002) 323.
02Sin Singh, I.S., Suri, M.M.K., Vidhani, J.M., Garg, S.P., Sharma, R.C.: Radiat. Prot. Dosim. 102
(2) (2002) 145.
03Doe Doerfel, H., Andrasi, A., Bailey, M.R., Berkovski, V., Castellani, C.-M., Hurtgen, C., Jourdain,
J.-R., LeGuen, B.: Radiat. Prot. Dosim. 105 (1-4) (2003) 645.
03Hun Hunt, J.G., de S. Santos, D., da Silva, F.C., Malatova, I., Foltanova, S., Dantas, B.M., Azaredo,
A.: Radiat. Prot. Dosim. 105 (1-4) (2003) 549.
04BMU Bundesministerium fr Umwelt, Naturschutz und Reaktorsicherheit: Richtlinie zur Ermittlung
der Krperdosis bei innerer Strahlenexposition, Bundesanzeiger, to be published (2004).

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-97]

10 Measuring techniques

10-83

10.3.3 In vitro measurements: excretion analyses


10.3.3.1 Introduction
In section 7.5.1.2 Analysis of excreta and other biological materials the general recommendations of the
ICRP in its Publication 78 [78ICR] on excreta monitoring programmes are summarized.
The selection, use and interpretation of various bioassay approaches are based on the physical and
biokinetic characteristics of the particular radionuclides considered. In vitro analysis refers to the
identification and quantification of radionuclides in the body by analysis of material excreted or removed
from the body. The main sources of bioassay data are urine, faeces, breath and blood. Other samples such
as hair, teeth, saliva and nails have been employed in special cases. These biological samples provide an
indirect measure of the internal radionuclide deposition because there is no direct information about the
body or organ burdens. Proper interpretation of these results requires knowledge of the relationship
between the presence of a radionuclide in the various bioassay samples and the organ radionuclide
burdens of interest.
Various factors influence the applicability of any particular type of sample:

The chemical element involved,


Its physical and chemical form,
The magnitude of internal deposition,
Biological and physical half-lives of the radionuclides involved,
Time elapsed since the intake occurred, and
Sensitivity of the analytical and measuring method used.

For radionuclides emitting non-penetrating radiation, i.e. radiation being absorbed in the body,
excretion analysis is the common method for monitoring workers exposed to radioactive material. This is
the case with radionuclides with alpha particle radiation (such as thorium, uranium, plutonium,
americium, curium) and radionuclides with soft beta particle radiation (3H, 14C, 35S, and others).
In vitro techniques for evaluating the internal contamination include also time-consuming procedures
for processing urine or faeces samples. Therefore, this technique should be used when in vivo methods are
not applicable and the exposure is at low levels.
A critical point of the in vitro analysis is the sampling. For the application of common assessment
models the excretion samples have to be collected in a well defined time interval. Another important point
is that the collected bioassay samples have to be free of contaminations from outside.
For the interpretation of the data the biological variability of the excretion of a person, which can lead
to different fractions of total body activity to be excreted in daily samples, must be considered.
Consideration should be also given to whether medical interventions (chelating therapy, administration of
diuretes, blocking agents and so on) could have influenced the pathway or excretion rates of
radionuclides.
10.3.3.2 Urine samples
Analysis of urine samples for excreted radionuclides is the method used most frequently for routine
monitoring and assessment of internal contamination. Urine samples are easy to collect and rather reliably
interpreted for material readily absorbed in the gastrointestinal (GI) tract.
A radionuclide in a relatively transferable (soluble) form entering the body reaches the bloodstream
and a fraction of it is deposited in various body organs. The remainder is excreted predominantly in the
urine. This biokinetic behaviour depends on the chemical form of the radionuclide involved entering the
body, and from its metabolic behaviour in the body after incorporation. Typical radionuclides which will
be monitored routinely via urine samples are for example 3H, 32P, 35S, 89Sr, 147Pm, thorium, americium
and other alpha-emitting radionuclides (see also Table 10.24).

Landolt-Brnstein
New Series VIII/4

10-84

10 Measuring techniques

[Ref. p. 10-97

The nominal daily excretion rate of urine amounts to 1.2 l for females and 1.6 l for males [89ICR].
The individual excretion rate depends strongly on physiological and environmental conditions but also on
individual nutritional habits. Therefore, the general sampling practice for routine monitoring is to collect
24-h-urine samples or equivalent. Repetitive sampling helps determining the time-dependent rate of
excretion of a radionuclide after intake of the radionuclide into the body.
Radioactive material can be lost from solution by adsorption onto surfaces of some containers, and so
on. For this reason samples must often be stabilised until analysis by refrigeration or freezing. Other
methods are the addition of a carrier or of an acidic, basic or other preservative as is appropriate for the
particular situation.
10.3.3.3 Faeces samples
Collection and analysis of faecal samples is another means of obtaining an indirect assessment of possible
internal contamination. For routine monitoring, faeces are not used as often as urine, but analysis of
faeces can provide at least qualitative information, particularly for relatively insoluble radionuclides.
Faeces samples are also very helpful for the quick assessment in the case of an extraordinary situation:
The excretion of radionuclides by faeces specifically of those with low gastrointestinal absorption (see
Chapter 7) is very often faster than by urine.
When an intake occurrs by ingestion, the quantity of a radionuclide being excreted soon after
ingestion represents the fraction of the radionuclide that has not been absorbed during the passage through
the GI tract. In the case of inhalation there are two fractions; one fraction which is absorbed from the
respiratory tract enters the bloodstream and is partly deposited in various organs. A part of it is
subsequently excreted from the liver into the GI tract via the bile into the faeces. The second contribution
comes from radionuclides translocated by swallowing from the respiratory tract directly into the GI tract
and is partly retained from the small intestine or excreted via the faeces (see Chapter 7).
Long-term excretion of a radionuclide by faeces after its intake into the body is originating from
delayed clearance of insoluble material from the pulmonary region of the respiratory tract or from the
clearance of material that has entered the bloodstream and is excreted from the liver into the GI tract via
the bile.
There are only few radionuclides for which a routine monitoring should be based on faecal excretion
analysis: 90Y, 147Pm (inhalation type S), thorium, curium and other alpha-emitting radionuclides (see also
Table 10.24). Such a monitoring is adequate to identify incorporations which occurred just before
sampling. Additionally, annual or biannual monitoring by faecal sampling may be used to check the
reliability of air monitoring. On the basis of air monitoring results of a hypothetical faecal excretion rate
can be derived; the comparison of measured faecal analysis can help to exclude a significant
underestimation.
The general sampling practice for routine monitoring is to collect 24-h-faecal samples. The nominal
transit time for material passing directly through the GI tract is about two days [30ICR], but this varies
considerably with diet, health of the individual, and other factors. For this reason also a collection time of
three consecutive days for faecal samples is recommended, in order to obtain reliable estimates of daily
excretion rates.
Faecal samples are particularly subject to biodegradation. Therefore, they should be analysed
promptly, ashed or preserved by deep freezing.
10.3.3.4 Exhalation
Breath samples can be useful in the case of incorporation monitoring for determining the amount of a
radionuclide leaving the body by exhalation, i.e. in gaseous form such as 222Rn, 220Rn, 14C-labelled carbon
dioxide or tritiated water vapour.
In the case of tritiated water about one third of an intake is excreted via breath, whose specific activity
rapidly reaches equilibrium with that in body water [00IAE].

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-97]

10 Measuring techniques

10-85

For example, 220Rn exhalation measurement [00Eis] allows the individual determination of 228Th body
burdens without chemical preparation. That means, 228Th will be measured by its decay product 220Rn
(daughter of 224Ra) in the exhaled air of a person with thorium burden. So the worker has for breath
sampling only to breathe into a collecting apparatus for up to 30 minutes, depending on the volume
required. The detection limit of this method is about 1 Bq of 228Th in the lung and thus comparable to that
of urinary excretion analysis. So this exhalation measurement method is best used to complement other
assessment methods.
Quantification of 232Th by measurement of 220Rn in exhaled air requires additional information about
the nuclide spectrum because of the unknown activity ratio between 228Th and 232Th in the body. For
example, the diet and the mineral water could lead via 228Ra to an increased 228Th burden.
10.3.3.5 Other biological samples
Other biological samples, such as blood, hair, teeth, nails and nose blows can be used only as indicators
for intake of radionuclides. Due to lack of biokinetic models or uncertainties in those models and data
used for qualitative assessment of any internal contamination the results of assessments are generally not
useful as a basis for quantitative dose estimations.
Blood samples provide the most direct source for estimating circulating internal contamination. But
the majority of radionuclides are rapidly cleared from the blood. So, and because of recirculation in the
body, measurements of activity in blood are generally only poor indicators of the total systemic content.
The analysis of nose blow samples can supplement monitoring for the purpose of screening for
intakes, and give valuable information on the nature of the inhaled contaminant. Usually this method
triggers other types or complementary analyses, such as urine or faeces samples.
Hair samples have been analysed for plutonium [81Too]. Caution is needed to ensure that hair care
products have not resulted in contamination with naturally occurring radionuclides, such as uranium.
Teeth incorporate many of the bone-seeking elements, such as strontium, and may provide an
indication of long-term, e.g. childhood exposures [95Hen].
10.3.3.6 Radiochemical analyses
A large number of different in vitro techniques have been developed for the detection and quantification
of low-level activity of radionuclides in excreta. Table 10.24 gives an overview on some typical
radionuclides and the complexity of the analytical procedure needed.
Table 10.24.
F = faeces)
Radionuclide
3
H
14
C
32
P
90
Sr
131

Examples for some typical radionuclides for monitoring by in vitro techniqes (U = urine,
Inhalation type,
Chemical compound
HTO
org.
F, M
F, S

I
Pm
232
Th

F
M
M, S

239

147

Pu

Landolt-Brnstein
New Series VIII/4

Biological sample

Analytical procedure

U
U
U
U
F
U
U
U
F
U
F

simple
simple
simple
elaborate
elaborate
simple
elaborate
elaborate
elaborate
elaborate
elaborate

10-86

10 Measuring techniques

[Ref. p. 10-97

Explanation
Radionuclide:
Inhalation type:

The radionuclide possibly suitable for incorporation monitoring.


Inhalation type of the appropriate radionuclide specified as
F = fast lung absorption,
M = moderate lung absorption, and
S = slow lung absorption.
Biological sample:
Determination of the activity in urine (U) samples or in faeces (F) samples.
Analytical procedure: A rough overview on the extent of the analytical procedure (simple, elaborate) necessary to determine the radionuclide in the appropriate sample.

In general it is possible to classify the procedure for the determination of the activity concentration of
the radionuclides in the following four main groups:
Alpha particle spectrometry of urine samples, using elaborate and time consuming radiochemical procedures, e.g., for 232Th, 241Am, 239Pu;
Beta counting of urine samples without or with simple radiochemical procedures, e.g., for 3H, 14C, 32P;
Beta counting of urine or faecal samples using elaborate and time consuming radiochemical procedures, e.g., for 90Sr and 147Pm; and
Gamma counting of urine samples without or with simple preceding radiochemical procedures, e.g.,
for 60Co, 134Cs, 137Cs and 131I.
Regardless of which in vitro technique is used, the sample activity is calculated after the measurement
by the equation

Ac = (

N s Nb 1 1 1

)
ts
tb
R E V

(10.3.3.1)

where
Ac = sample activity concentration in Bq per volume unit
Ns = number of counts observed in the sample during the counting time ts
Nb = number of counts of the background during the counting time tb
ts = sample counting time
tb = background counting time
R = chemical recovery, expressed as a fraction
E = counting efficiency, expressed as a fraction
V = sample size as volume unit
If the counting times are equal the equation above will reduce to

1
1 1 1
A c = (N s N b )
t
R E V

(10.3.3.2)

When an internal tracer is added, e.g., in the case of alpha particle spectrometry, the activity may be
calculated using this equation

N N
b
A = At s
N t N b'

(10.3.3.3)

Where
A = sample activity
At = activity of the internal tracer added to the sample
Nt = number of counts observed in the tracer region of interest (ROI)
Nb = number of counts of the background in the tracer ROI
K = calibration factor applicable for the sample volume

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-97]

10 Measuring techniques

10-87

In this case it is not necessary to know the chemical recovery and the efficiency.
The minimum detectable activity (MDA) corresponds to the level of activity which is required to
ensure with some chosen level of confidence that the net signal will be detected. The definitions and
equations are specified in the Safety Standards Series No. RS-G-1.2 [99IAE] and Safety Reports Series
No. 18 [00IAE]. See also Section 10.3.2.8.

MDA = 3.3

Nb
ts

t 1 1 1
1 + s
tb R E V

(10.3.3.4)

When , the probability of a type I error (false positive), and , the probability of a type II error (false
negative), are both set equal to 0.05, MDA may be calculated in most cases as shown above (the symbols
are the same as in the equations before).
In excretion analysis samples usually contain only low activities of a radionuclide. To ensure the
reliable detection of such small activities low detection limits are essential. Therefore the following
technical items have to be taken into consideration:
The radiochemical recovery must be as high as possible.
In general the counting or detection efficiency for alpha or beta particles cannot be higher than 50 %
in the case of measuring flat source discs, because the solid angle seen by the detector cannot be more
than 2. On the other hand the counting efficiency is a function of the distance between detector and
source and varies for alpha particle spectrometry from a few percent up to 40 %. Therefore it is
necessary to get an optimal adjustment between counting efficiency and energy resolution needed.
A higher volume of the sample leads to a lower MDA per volume unit (see equation (10.3.3.4)).
A lower number of background counts will result in a lower MDA. A main point to ensure this
requirement is the careful radiochemical separation of the radionuclides which have to be
determined.
The MDA is inversely proportional to the counting time. That means the sample counting time should
be as long as possible, but it depends on the time available.
Equations for the calculation of MDA for more complicated cases will be found in the Safety Reports
Series No. 18 [00IAE].
Whatever technique is used for counting, most of the radionuclides analysed and especially the
actinides, need to be isolated from the matrices. Numerous analytical procedures have been developed.
They all are based on the same principle which consists of
Sample preparation,
chemical separation, and
source preparation.
The sample preparation for urine involves wet ashing or co-precipitation of calcium phosphate,
calcination and dissolution of the precipitate. Faeces are ashed and dissolved in acid. Insoluble materials
such as silica are treated with fluorhydric acid. Similar procedures are used for tissue samples.
The chemical separation of the radionuclide to be analysed includes separation and purification on
ion-exchange resin or by solvent extraction, or a combination of these two techniques.
The type of source preparation used is a function of the following measuring technique needed for the
analysis.
For the alpha particle spectrometry of actinides the source is prepared by
Direct evaporation,
co-precipitation with lanthanide fluoride, or
electrodeposition.
Landolt-Brnstein
New Series VIII/4

10-88

10 Measuring techniques

[Ref. p. 10-97

For beta counting with a proportional counter, the source is generally obtained by precipitation and
filtration of the insoluble salt. For liquid scintillation counting (LSC) the purified radionuclide solution is
mixed with an appropriate cocktail for the measurement.
The radiochemical analyses carried out routinely in the laboratory for monitoring potential
incorporations of occupationally exposed workers has to be documented [00ISO2]. These written
procedures shall include all steps starting with the receipt of the sample at the laboratory to measurement
of the sample, or of an aliquot of the sample, and should contain all radiochemical procedures used.
For example, the description of the procedures can be very different:
In the simple case of analysing tritium (see Table 10.24) it is only necessary to describe how to make
the aliquot of the urine sample and the adding of an appropriate scintillation substance to be ready for
the measurement by liquid scintillation counter.
But in most cases the analytical procedure is more complicated and time-consuming, as seen in Table
10.24. In connection with the sample preparation (urine, faeces, tissue, blood etc.) and after adding an
internal tracer to determine the chemical recovery, several nuclide-specific radiochemical separation
steps have to be done. After electrodepostion on a stainless steel disk the radionuclide activity in the
sample can be determined, e.g., by alpha particle spectrometry.
A lot of different analytical procedures have been published. A very fundamental publication in this
field is the HASL-300 (Health and Safety Laboratory) document [97HAS]. It is well known as The
procedure manual of the Environmental Measurements Laboratory (EML) and covers the existing
technology and procedures currently in use at EML (older procedures are updated and new procedures are
added). This voluminous manual is also available as CD-ROM. The main task of this document is the
analytical chemistry to be used for a wide range of radionuclides (e.g., 3H, Fe, Sr, Tc, Pb, Po) with
different matrices and measuring techniques. Detailed descriptions for the determination of thorium in
urine and faeces, used in different laboratories in Germany, are given by Riedel et al. [93Rie]. The
contribution of Harduin et al. [96Har] describes the analytical determination, especially for actinides in
biological samples. Instructions about sequence analysis of actinides and 90Sr are found in the work of
Wihlidal et al. [98Wih]. A fundamental and very informative technical note about the electrodeposition of
actinides is given by L. Hallstadius [84Hal].
An important application of these analytical procedures and measuring techniques is the monitoring of
workers involved in the decommissioning of nuclear facilities, because of the great variety of the
radionuclides present and the conditions of the exposure. Establishing appropriate monitoring programmes and procedures is currently in progress. Due to the presence of transuranic radionuclides the
analysis of excretion samples is required. So, for example, the contribution of Robredo et al. [00Rob]
describes an excellent radiochemical procedure for the determination of americium and plutonium in
urine samples. Neudert et al. [99Neu] present a very fast and closely method by using inductively coupled
plasma-mass spectrometry (ICP-MS).
In case of monitoring potential incorporations of occupationally exposed workers, in some cases
consideration has to be given to radionuclides incorporated from natural sources via food, especially
drinking water. These intakes result in contributions of activities measured when monitoring workers and
may mislead the dosimetry of occupationally exposed workers. Therefore it is necessary to have
information on the natural contents, especially of thorium and uranium in human urine and faeces; see for
example [97Rot] and [98Bey].
10.3.3.7 Measuring techniques
There exists a wide field of different measuring techniques to determine the activity of alpha and beta
emitting radionuclides (alpha particle spectrometry, beta counting, liquid scintillation counting (LSC),
fluorimetry, laser induced fluorimetry, gamma spectrometry, inductively coupled plasma-mass
spectrometry (ICP-MS), neutron activation analysis (NAA) and delayed neutron activation analysis
(DNAA)).

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-97]

10 Measuring techniques

10-89

Table 10.25 gives a rough overview of these different techniques for the determination of
radionuclides in excreta, including the values for the minimum detectable activity (MDA). The main
measuring techniques for the determination of radionuclides in excretion samples are summarised below.
Alpha particle spectrometry
This technique is the most commonly applied technique for measuring the isotopes of the different alpha
emitters, such as thorium, uranium, plutonium, americium and curium. Alpha particle spectrometry is
used to identify the isotopes and to quantify their activities.
Because of the non-penetrating radiation of the alpha emitter, the alpha particle spectrometry requires
prior to the measurement an elaborate and time-consuming radiochemical procedure, as described above.
For a quantitative nuclide-specific separation a very thin source is very important to get a good energy
resolution of the alpha particle spectrum. So the common source preparation technique used is the
electrodeposition on a stainless steel disk. This flat source will be measured in an alpha chamber in
connection with a multichannel analyser to determine the different isotopes [84Hol].
By using internal tracers such as 229Th, 232U, 242Pu and 243Am the radiochemical recovery is simple to
determine. This also allows to measure all isotopes of the element analysed in the same spectrum with the
same MDA.
Beta counting
Radionuclides such as 32P, 89Sr, 90Sr or 131I can be measured after the chemical separation with a low
background proportional counter. The technique is applicable for beta emitters of relatively high energy.
As for alpha particle spectrometry, this technique requires the same type of radiochemical procedure as
described before. The preparation of the source is generally obtained by precipitation and filtration of the
unsoluble salt.
Table 10.25. Measuring techniques for the analyses of excreta
Measuring
Radionuclide
Analytical
technique
procedure
Alpha particle
Th, U, Pu, Am, Cm
elaborate
spectrometry
32
Beta counting
P, 89Sr, 90Sr
elaborate
3
LSC
H, 14C, 63Ni,
no
241
Pu
elaborate
32
P, 89Sr, 90Sr
elaborate
60
Gamma
Co, 134Cs, 137Cs
no
spectrometry
Fluorimetry
U nat
simple
Laser-induced
U nat
simple
fluorimetry
232
ICP-MS
Th,
no
238
U,
no
239
Pu,
elaborate
240
Pu
elaborate
238
NAA
U,
elaborate
232
Th
elaborate
235
DNAA
U,
no
U nat,
no
U nat
simple

Landolt-Brnstein
New Series VIII/4

Typical
MDA
1.0 mBq/l
<80 mBq/l
5 Bq/l
15 mBq/l
120 mBq/l
200 mBq/l
25 mBq/l
2 mBq/l
0.05 mBq/l
0.1 mBq/l
0.1 mBq/l
0.4 mBq/l
0.0025 mBq/l
0.1 mBq/l
60 mBq/l
25 mBq/l
0.5 mBq/l

10-90

10 Measuring techniques

[Ref. p. 10-97

Measurement with a low background proportional counter will lower the MDA as compared to
measurement by liquid scintillation counting (LSC).
Liquid scintillation counting is especially useful for beta emitters of weak energy such as 3H, 14C and
63
Ni. For these radionuclides a direct measurement can be performed by mixing a small volume of the
urine sample with the scintillation cocktail. Internal standards are often used to measure low-level
radioactivity.
Liquid scintillation counting is also used to determine pure beta emitters like 32P, 89Sr and 90Sr. For the
measurements of these radionuclides an elaborate radiochemical procedure is needed for separating the
analyte from the matrix.
Fluorimetry
Fluorimetry is a simple and fast technique for the determination of uranium in urine. Uranium is
determined by the fluorescence produced when exposed to ultraviolet light. The urine sample may
directly be fused in a platinium dish and measured with a fluorimeter. The limitation of this technique is
its poor sensitivity and the measurement of total uranium only, mainly 238U.
Laser-induced fluorimetry
The laser-induced fluorescence excitation technique needs some treatment of the urine like co-precipitation and calcination, or wet ashing before measuring the uranium content.
Gamma spectrometry
In many cases gamma spectrometry can be directly applied on the urine samples. It should be noticed that
for gamma emitters such as 60Co, 134Cs, 137Cs and 131I in vivo measurements are mainly performed.
Inductively coupled plasma-mass spectrometry (ICP-MS)
The sample is introduced into a mass spectrometer by means of a nebuliser in a plasma torch, and
afterwards in a magnetic field that separates the atoms in their different masses and producing thus results
for each of the isotopes of the element analysed. This technique has been used for uranium and thorium
analyses in urine and human tissues. In fact, this technique is very sensitive for radionuclides with very
long half-life such as 235U, 238U and 232Th. Beside acidifying the urine sample no other treatment is
needed.
This mass spectrometry is also a very sensitive technique for measuring isotopic composition of
plutonium. For plutonium analysis a radiochemical procedure is needed to separate plutonium from the
bulk of inorganic material and from 238U which causes interferences at the mass 239, before being
subjected to the mass spectrometric analysis. This technique is capable of measuring 239Pu and 240Pu
separately, which is not possible by alpha particle spectrometry.
A clear advantage of ICP-MS is its rapidity. After treatment of the sample, if needed, the results can
be obtained in a few minutes; see for example [98Kre], [02Sch] and [03Bou].
Neutron activation analysis (NAA)
The neutron activation analysis has been used for the rapid determination of thorium in urine and in
biological fluids, like serum. The technique involves some radiochemical procedures before and after
irradiation of thorium with thermal neutrons, see for example [94Hub]. The photons of 233Pa obtained by
irradiation of 232Th are counted. This technique is sufficiently sensitive to detect the natural content of
232
Th in the environment, especially in food.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-97]

10 Measuring techniques

10-91

The NAA technique has also been reported for the determination of 238U in urine. In this case the
photons of 239Np will be counted.
Delayed neutron activation analysis (DNAA)
Delayed neutron assay has been used for measuring 235U. As a screening method for uranium in urine,
the evaporated urine was directly irradiated by thermal neutrons in a nuclear reactor. Then the delayed
neutrons resulting from fisson of uranium were counted. Good compilations about this measuring
technique are found in [84Gab], [89Gla] and [94Hub]. For the determination of uranium in urine there is
nearly no sample preparation necessary.
232
Th could be fisured only by epithermal neutrons. For the analysis of thorium in urine samples a
radiochemical separation technique is required.
DNAA is a very fast technique but measures only 235U, requiring information concerning the isotopic
composition of uranium. For natural uranium a lower MDA can be achieved if uranium is isolated by
adequate radiochemical separation.
10.3.3.8 Quality assurance
Quality assurance (QA) is an essential and integral element of the routine work in a laboratory to ensure
the reliability of the bioassay data yielded in this laboratory. The QA practice in the laboratory can be
achieved, e.g., by:
Establishing a certified quality management system
Accreditation of all analytical procedures and measuring techniques used in the routine work of this
laboratory.
The normative requirements on a certified quality management system are laid down in EN ISO 9001:
2000 [00ISO1]. For the accreditation normative requirements exist for testing laboratories (i.e. producing activity values) in EN ISO/IEC 17025: 2000 [00ISO2]. In the case of laboratories performing
inspections (i.e. producing activity values including interpretation, e.g., dose values) normative criteria
are specified in ISO/IEC 17020: 1998 [98ISO]. General and helpful support is given in the paper Quality
management systems for technical services in radiation safety [03IAE].
The laboratory has to establish an in-house quality assurance plan to prove its organisational and
technical competence. This plan should consist of two sections: the more formal part of administration
support, and the technical part of quality control. In the first section the plan should include, among
others, the following items:

Organisational responsibilities
Corrective actions
Personnel qualifications
Adequate operational environment
Documentation of all aspects of the bioassay monitoring programme.
The technical part of the QA plan should include procedures as follows:

Sample registration procedures


Administrative procedure for each sample
Procedure manuals for the radiochemical analyses and measuring techniques
Instrumentation and calibration manuals
Use of control charts and testing materials
Participation in intra- and interlaboratory comparisons.

Landolt-Brnstein
New Series VIII/4

10-92

10 Measuring techniques

[Ref. p. 10-97

More detailed instructions are given in IAEA Safety Reports Series No. 18 [00IAE] and in ISO
12790-1 [01ISO]. The design and implementation of a QA plan is described in the Safety Guide No. RSG-1.2 [99IAE].
In order to avoid systematic errors and ensure the quality of the analyses and techniques used, many
additional measurements and methods have to be performed in the laboratory. For example, routine
checks are necessary on:

Radiochemical recovery
Energy calibration
Efficiency calibration
Background measurements
Blind analyses (blanks).

Based on the laboratory practice of many years, particularly the following problems have to be
considered.
Uncritical observance of analysis procedures
Generally analysis procedures are given for a defined matrix, e.g. for urine samples with an average
content of mineral salt. In practice, however, the salt content is occasionally very high, e.g. from the
administration of calcium tablets or excess consumption of lemonades with high phosphate content. In
this case procedures should be modified by introducing additional separation steps or by using more
efficient separation methods.
Avoiding cross-contamination
Cross-contamination, particularly in monitoring incorporations of alpha emitters, may lead to error
analyses with serious consequences. They occur when samples with very different concentrations are
measured with the same laboratory equipment. When uranium and thorium isotopes are determined it is
appropriate to conduct the measurement of urine and faeces samples in separate locations and with
different equipment, because their natural excretion is quite different.
Errors during internal standardisation
In the course of internal standardisation being commonly used in radiochemical analyses, the standard
may contribute to the lasting inaccuracy of whole series of measuring results. This may be caused from,
among others, errors of production or inappropriate storage and application. In some cases a cleaning of
the standard from daughter nuclides is required immediately before it is used for analysis procedures.
Interference due to blind values from chemicals
To assess natural 232Th excretions in urine, identification limits of a few Bq excretions per day are
required. In this range even p.A qualities of the used chemicals may lead to relevant contamination. Thus
in concentrated hydrochloric and nitric acid (each at p.A. quality) up to a maximum of 3 mBq 232Th per
litre have been proved.

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-97]

10 Measuring techniques

10-93

10.3.3.9 Examples for dose estimations from in vitro measurements


Case 1: Routine monitoring of depleted uranium (238U)
Handling: Depleted uranium, ICRP inhalation type M,
routine monitoring by analysis of the urine,
two times per year (every 180 days)
Measured value m = 21 mBq/d (24 h urine)
Dose assessment by using the following standard assumptions:

Route of intake: acute inhalation


Date of incorporation: in the middle of the monitoring interval
AMAD: 5 m
Biokinetic and dosimetric data: ICRP 68 and ICRP 78.

The committed effective dose is calculated by the equation:


E=

e(50) m
E ( t )
E

(10.3.3.5)

where
E
= committed effective dose in Sv
e(50) = dose coefficient for the effective dose in Sv/Bq
m
= value of the 24 h excretion in Bq/d, corrected at the end of the collecting period
EE (t) = excretion rate (EE = EU for urine), at the day t in Bq/d after acute inhalation of 1 Bq
t
= in the case of dose assessment by using standard assumptions half-time of the monitoring
interval in days
Result:
Committed effective dose E = 0.28 mSv half-yearly, calculated for 238U
with
EU(t) = 1.2 104 Bq/d Bq1 at t = 90 days for 238U, tabulated in [03Nos]
e(50) = 1.6 106 Sv/Bq for 238U, tabulated in [78ICR]
Case 2: Practical estimation of the date of incorporation of 35S
Handling:

35

S, ICRP inhalation type F,


routine monitoring by analysis of the urine monthly, i.e. every 30 days
Data from the 1st monitoring measurement:
Date of collecting the urine: 30.04./01.05.2001, 24 h urine.
Collected urine sample: 1100 ml, measured value 37 Bq/5 ml urine.
Measurement result m = 8.1 kBq/d
Dose assessment by using the same standard assumptions as in case 1, but intake and committed effective
dose will be calculated in two steps, derived from the equation (10.3.3.5) in case 1:
intake I = m/ EU(t ) = 7.4 MBq
and
committed effective dose E = I e (50) = 0.59 mSv monthly
with
EU (t ) = 1.1 103 Bq/d Bq1 at t = 15 days, tabulated in [03Nos]
e (50) = 8.0 1011 Sv/Bq for 35S, tabulated in [03Nos]

Landolt-Brnstein
New Series VIII/4

10-94

10 Measuring techniques

[Ref. p. 10-97

Problem: Notice of a significant content of 35S in the urine of one worker, but there was no remarkable
irregularity (contamination, accident and so on) happened in the laboratory during the monitoring
interval.
Therefore further measurements were done to try an estimation of the date of the incorporation and the
amount of the intake to get, on this basis, a more realistic effective dose value:
Collecting date Amount of urine Collecting time Measured value Remarks
[ml]
[h]
[Bq/5 ml urine]
30.04./01.05.01 1100
24
37
1st montoring
03.05./04.05.01

1300

24

2.1

special

07.05./08.05.01

600

12

2.1

measurements

02.06./03.06.01

1050

24

0.75

2nd monitoring

To obtain the date of incorporation and the amount of the intake of 35S the following information of
the radiation protection officer were used for the interpretation:
acute inhalation, and
inhalation type F and AMAD 5 m of the 35S compound involved.
The procedure consists of the following steps:
1. Variation of the date of the incorporation.
2. Calculation of the belonging intake for each of the four measured values.
3. Calculation of the mean intake and the standard deviation.
The mean value with the minimum standard deviation SD should be the realistic time of incorporation.
1.
2.
3.
4.
Mean
SD*)
Measurement Measurement Measurement Measurement value
Measured value 8140 Bq/d
546 Bq/d
504 Bq/d
158 Bq/d
27.04.2001:
4d
7d
11 d
36 d
Time t
1
1
1
0.0019 d
0.0016 d
0.0014 d
0.0005 d1
EU (t )
4284 kBq
341 kBq
360 kBq
316 kBq
1325 kBq 130 %
I = m/ EU (t )
28.04.2001:
3d
6d
10 d
35 d
Time t
0.0035 d1
0.0017 d1
0.0014 d1
0.0005 d1
EU (t )
2326 kBq
321 kBq
360 kBq
316 kBq
831 kBq
104 %
I = m/ EU (t )
29.04.2001:
2d
5d
9d
34 d
Time t
1
1
1
0.028 d
0.0017 d
0.0015 d
0.0005 d1
EU(t )
291 kBq
321 kBq
336 kBq
316 kBq
316 kBq
5.1 %
I = m/ EU(t )
30.04.2001:
1d
4d
8d
33 d
Time t
0.29 d1
0.0019 d1
0.0015 d1
0.0005 d1
EU (t )
28 kBq
287 kBq
336 kBq
316 kBq
242 kBq
52 %
I = m/ EU (t )
*) standard deviation

Landolt-Brnstein
New Series VIII/4

Ref. p. 10-97]

10 Measuring techniques

10-95

Result of the measurements:


The date of incorporation is 29.04.2001, as shown in the table above (the standard deviation is minimal at
this date).
The mean value of the intake I = 320 kBq.
The committed effective dose E = 0.25 mSv
Using this practical procedure in many cases it is possible to get infomation about the date of
incorporation (as shown in the example above), the route of intake and the value for the AMAD.
Case 3: Accidental inhalation of 239Pu [99Kau]
Situation: A worker inhaled contaminated aerosol for aproximately 10 minutes, 239Pu-oxide is involved
The following measurement results were achieved:
Time after the incident
[d]
1
2
3
10
15

Faecal activity
[Bq]
160
0.62
0.44

Urinary activity
[mBq]
1.6
1.1
-

Each measurement value will be interpreted in terms of intake using the ICRP Publication 78
[78ICR]:
The first faecal measurement was a measurement of the pool of all faecal excretion during the first
three days after the incident. To interpret this it is necessary to compare this value with the sum of the
parameters for the first three days given in [78ICR].
Unfortunately for 15 days no value is tabulated in [78ICR]. However, in the table for routine monitoring the value for the measurement period 30 days is suitable for this purpose because for routine
monitoring an acute intake in the middle of the monitoring interval is assumed, i.e. 15 days before the
measurement for a monitoring interval of 30 days.
Because 239Pu has been inhaled as an oxide, inhalation type S is assumed.
With these assumptions the following intake values were derived, using equation (10.3.3.5):
for fecal excretion measurements
I1 = m / [EF (1) + EF (2) + EF (3)] = 452 Bq
I2 = m / EF (10) = 954 Bq
I3 = m / EF (15) = 898 Bq
for urinary excretion measurements
I4 = m / EU (1) = 696 Bq
I5 = m / EU (3) = 1320 Bq

Landolt-Brnstein
New Series VIII/4

10-96

10 Measuring techniques

[Ref. p. 10-97

Using the following data for EF (t ) and EU (t ), tabulated in [78ICR]:


Time after intake
Urinary excretion
Faecal excretion
[d]
[Bq/d Bq1]
[Bq/d Bq1]
6
1
1.1 101
2.3 10
2
1.6 101
3
8.3 107
8.4 102
10
6.5 104
)
15*
4.9 104*)
*) Data for routine monitoring with an time interval of 30 days
Results:
In this case urinary and faecal excretion analyses give similar results. If several successive results are
available, the best estimation of the intake I is obtained by taking the geometric mean of the Ii values
established from these measurements. So in this case the geometric mean of all intake values is 810 Bq.
Committed effective dose E = 6.7 mSv
with
e(50) = 8.3 106 Sv/Bq for 239Pu, tabulated in [78ICR]
In the case that there would have been an excretion enhancement by DTPA it would be necessary to
consider this by some modifications, for example by divison of the measurement values by an appropriate
number.

Landolt-Brnstein
New Series VIII/4

10 Measuring techniques

10-97

10.3.3.10 References for 10.3.3


30ICR
78ICR
81Too
84Gab
84Hal
84Hol
89Gla
89ICR
93Rie
94Hub
95Hen

96Har
97HAS
97Rot
98Bey

98ISO
98Kre

98Wih
99IAE

International Commission on Radiological Protection: Limits for intakes of radionuclides by


workers; ICRP Publication 30, Oxford: Pergamon Press, 1979.
International Commission on Radiological Protection: Individual monitoring for internal
exposure of workers; ICRP Publication 78, Oxford: Pergamon Press, 1997.
Toohey, R.E., Cacic, C.G., Larsen, R.P., Oldham, R.D.: The concentration of plutonium in
hair following intravenous injection; Health Phys. 40 (1981) 881.
Gabelmann, H., Lerch, M., Kratz, K.-L., Rudolph, W.: Determination of uranium in urine
samples of fuel element fabrication workers by beta-delayed neutron counting; Nucl. Instrum.
Methods Phys. Res. 223 (1984) 544.
Hallstadius, L.: A method for the electrodeposition of actinides; Nucl. Instrum. Methods
Phys. Res. 223 (1984) 266.
Holm, E.: Review of alpha-particle spectrometric measurements of actinides; Int. J. Appl.
Radiat. Isot. 35 (4) (1984) 285.
Gladney, E.S., Moss, W.D., Gautier, M.A., Bell, M.G.: Determination of U in urine:
Comparison of ICP-mass spectrometry and delayed neutron assay; Health Phys. 57 (1)
(1989) 171.
International Commission on Radiological Protection: Basic anatomical and physiological
data for the use in radiological protection: Reference values; ICRP Publication 89, Oxford:
Pergamon Press, 2002.
Riedel, W., Beyer, D., Dalheimer, A., Doerfel, H., Henrichs, H., Scheler, R.:
Inkorporationsberwachung auf Thorium; Reihe: Fortschritte im Strahlenschutz, FS-93-69AKI, 1993.
Huber, G., Lenz, S., Pfeiffer, B., Kratz, K.-L.: Bestimmung von Uran und Thorium mittels
Neutronenaktivierungsanalyse; Universitt Mainz, Bericht IKMZ 94-5, 1994.
Henshaw, D.L., Allen, J.E., Keitch, P.A., Salmon, P.L., Oyedepo, C.: The microdistribution
of 210Po with respect to bone surfaces in adults, children and fetal tissues at natural exposure
levels; in: Health effects of internally deposited radionuclides: Emphasis on radium and
thorium, van Kaick, G., Karaoglou, A., Kellerer, A.M. (eds.) EUR 15877 EN, Singapore:
World Scientific Publishing, 1995, p. 23-26.
Harduin, J.C., Peleau, B., Levasseur, D.: Analytical determination of actinides in biological
samples; Radioprotection 31 (2) (1996) 229.
Environmental Measurements Laboratory: The procedure manual of the Environmental
Measurements Laboratory; HASL-300, volume I + II, 28th edition (1997); and as CD-ROM,
www.eml.doe.gov (2002).
Roth, P., Werner, E., Wendler, I., Schramel, P.: Variation of natural 232Th excretion in nonexposed persons; J. Radioanal. Nucl. Chem. 226 (1-2) (1997) 285.
Beyer, D., Dalheimer, A., Riedel, W., Neudert, N.: Die Bedeutung der natrlichen
Ausscheidung bei der Inkorporationsberwachung auf U-238, Th-232, Sr-90 und Ra-226; in:
Radioaktivitt in Mensch und Umwelt, Volume I, Winter, M., Henrichs, K., Doerfel, H.
(eds.), Verlag TV Rheinland GmbH, 1998, p. 210-215.
International Organization for Standardization: General criteria for the operation of various
types of bodies performing inspection; ISO/IEC 17020, 1998, (Genve 1998).
Krec, T., Neudert, N.: Eignung eines ICP-MS-Mesystems fr die Inkorporationsberwachung auf Actiniden durch Ausscheidungsanalyse; in: Radioaktivitt in Mensch und
Umwelt, Volume I, Winter, M., Henrichs, K., Doerfel, H. (eds.), Verlag TV Rheinland
GmbH, 1998, p. 51-55.
Wihlidal, H., Sinojmeri, M., Lovranich, E., Steger, F.: Sequence analysis of actinides and
Sr-90 in urine samples; in: Radioaktivitt in Mensch und Umwelt, Volume I, Winter, M.,
Henrichs, K., Doerfel, H. (eds.), Verlag TV Rheinland GmbH, 1998, p. 222-226.
International Atomic Energy Agency: Assessment of occupational exposure due to intakes of
radionuclides; IAEA Safety Standards Series No. RS-G-1.2, Vienna, 1999.

Landolt-Brnstein
New Series VIII/4

10-98
99Kau
99Neu
00Eis
00IAE
00ISO1
00ISO2
00Rob
01ISO
02Sch
03Bou
03IAE
03Nos

10 Measuring techniques
Kaul, A.: Radiation dose assessment: Internal dose; Lecture notes, IAEA Regional Training
Course in Radiation Protection, Johannesburg, S.A., 1999.
Neudert, N., Roth, W.: Improvement of incorporation monitoring during decommissioning of
the hot-cell facility at Karlstein; Proceedings of the 3rd European ALARA Network
Workshop on Managing Internal Exposures, 15.-18.11.1999, Neuherberg, 1999.
Eisenmenger, A.: Messung von Rn-220-Exhalation zur Ermittlung von strahlenschutzrelevanten Inkorporationen bei Beschftigten der Thorium-verarbeitenden Industrie und
Thorotrast-Patienten; Thesis, Freie Universitt Berlin, 2000.
International Atomic Energy Agency: Indirect methods for assessing intakes of radionuclides
causing occupational exposure; IAEA Safety Reports Series No. 18, Vienna, 2000.
International Organization for Standardization: Quality management systems - Requirements;
EN ISO 9001: 2000, CEN European Committee for Standardization, Brssel, 2000.
International Organization for Standardization: General requirements for the competence of
testing and calibration laboratories; EN ISO/IEC 17025: 2000, CEN European Committee for
Standardization, Brssel, 2000.
Robredo, L.M., Navarro, T., Sierra, I.: Indirect monitoring of internal exposure in the
decommissioning of a nuclear power plant in Spain; Appl. Radiat. Isot. 53 (2000) 345.
International Organization for Standardization: Radiation protection - Performance criteria
for radiobioassay -, part 1: General principles; ISO 12790-1, Genve, 2001.
Schramel, P.: Determination of 235U and 238U in urine samples using sector field inductively
coupled plasma mass spectrometry; J. Chromatogr. B 778 (2002) 275.
Bouvier-Capely, C., Baglan, N., Montgue, A., Cossonet, C.: Validation of uranium
determination in urine by ICP-MS; Health Phys. 85 (2) (2003) 216.
International Atomic Energy Agency: Quality management systems for technical services in
radiation safety; IAEA Working material, Vienna, 2003.
Noke, D., Dalheimer, A., Dettmann, K., Frasch, G., Hartmann, M., Karcher, K., Knig, K.,
Scheler, R., Strauch, H.: Retentions- und Ausscheidungsdaten sowie Dosiskoeffizienten fr
die Inkorporationsberwachung; BfS-Bericht BfS-SG-02/03, 2003.

Landolt-Brnstein
New Series VIII/4

Ref. p. 11-12]

11 Exposures from natural radiation sources

11-1

11 Exposures from natural and man-made radiation sources

Natural background form the baseline upon which all man-made exposures are added and against which
these exposures may be compared. Main contributors to the natural radiation exposure of man are
radiations of cosmic origin or radionuclides present in the earths crust including the human body itself.
Largely depending on altitudes above sea level, geomagnetic altitudes and geological external conditions
worldwide and internal doses from natural radiological sources vary from 1 to 10 mSv annually with an
average of 2.4 mSv a1. Nearly 50 % is from indoor inhalation of 222Rn and its progeny.

11.1 Introduction
The exposure of man to ionizing radiation from natural sources is a continuing and inescapable feature of
live on earth. For most induviduals, this natural background exposures are much more significant than the
exposures caused by man-made sources. Exceptions that apply to certain individuals are some exposures
caused by medical radiation procedures, through mishandling of radiation sources, in accidents allowing
radionuclides to be released to the environment, and at some workplaces. In all cases, the natural
background source form the baseline upon which all man-made exposures are added, and it is a common
level against which these exposures may be compared.
Essentially there are two main contributors to the natural radiation exposure of man: charged and
uncharged particles generated by high-engery particles of cosmic origin incident on the earth's
atmosphere, and radioactive nuclides originating either by interaction of cosmic-ray particles in the earth's
atmosphere or being naturally present in the earth's crust everywhere in the environment, including the
human body itself. From these sources dose to man arise from both external and internal exposure.
Exposure from extra-terrestrial sources (cosmic radiations and cosmogenic radionuclides) and of
terrestial origin (e.g. 40K, and radionuclides of the uranium and thorium decay chains) contribute to the
natural background at a comparatively constant level, although largely depending on geological
conditions, altitudes above sea level, and geomagnetic latitudes.
Origin and kinds of galactic and solar cosmic radiations and reference doses to members of the world
population arising from these external sources as well as internally from ingestion of cosmogenic
radionuclides are dealt with in Section 11.2. Exposure pathways and doses from terrestrial sources of
natural origin are described in Section 11.3 with special emphasis to the inhalation of radon progeny of
the uranium and thorium decay chains. In Section 11.4 exposures are summarized due to modification of
natural sources by human activities, e.g. release of natural radionuclides to the environment in mineral
processing and fossil fuel combustion. Finally, i.e. in Section 11.5, the worldwide average exposures from
the single sources of natural origin and the total dose by adding up the various components of the
effective dose are given together with the normal ranges of these exposures. These numbers serve as the
basis for evaluating the present (year 2000) doses from man-made sources of artificial origin and of
occupational radiation exposures.

11-2

11 Exposures from natural radiation sources

[Ref. p. 11-12

Quantitative information on sources and doses from natural and enhanced natural exposures due to
industrial activities as well as from exposures to the public from man-made sources of radiation such as
medical and occupational exposures, and those from peaceful and military defence uses of nuclear energy
is based on data recently published by the United Nations Scientific Committee on the Effects of Atomic
Radiation UNSCEAR in its 2000 Report [00U].

11.2 Exposures by cosmic radiation and cosmogenic radionuclides


11.2.1 Origin and kinds of cosmic radiation
Galactic cosmic rays incident on the top of the atmosphere consist of a nucleonic component (98 %), and
electrons (2 %). The nucleonic component is primarily protons (88 %) and -particles (11 %), with the
remainder heavier nuclei. These primary cosmic particles have an energy spectrum that extends from
108 eV to over 1020 eV.
Another component of cosmic rays is generated near the surface of the sun by magnetic disturbances.
These solar particle events are comprised mostly of protons of energies generally below 108 eV and only
rarely above 1010 eV. They can produce significant dose rates at high altitudes, but only the most
energetic affect dose rates of background level. Solar particle events are highly variable in intensity and
of short duration, typically a few hours. They have a negligible impact on long-term doses to the general
population. The most significant long-term solar effect is the 11-year cycle in solar activity, which
generates a corresponding cycle in total cosmic radiation intensity.
The magnetic field of the earth partly reduces the intensity of cosmic radiation reaching the top of
atmosphere. The form of the earth's field is such that only particles of higher energies can penetrate at
lower geomagnetic latitudes.
The high-energy particles incident on the atmosphere interact with atoms and molecules in the air and
generate a complex set of secondary charged and uncharged particles, including protons, neutrons, pions
and low-Z nuclei. The secondary nucleons in turn generate a cascade of more nucleons in the atmosphere.
Because of their longer mean free path, neutrons dominate the nucleonic component at lower altitudes.
The neutron energy distribution peaks between 50 and 500 MeV as well as around 1 MeV (produced by
nuclear deexcitation) are important in dose assessment.
The pions generated in nuclear interactions are the main source of the other components of the cosmic
radiation field of the atmosphere. The neutral pions decay into high-energy photons, which produce highenergy electrons, which in turn produce photons (photon/electron cascade). Electrons and positrons
dominate the charged particle fluence rate at middle altitudes. The charged pions decay into muons, the
dominant component of the charged-particle flux at ground level.

11.2.2 Exposures by cosmic radiations


At ground level, the muon component is the most important contributor to dose. At air craft altitudes,
neutrons, electrons, positrons, and photons are the most significant components. At higher altitudes, the
heavy nuclei components must by considered, too.

Ref. p. 11-12]

11 Exposures from natural radiation sources

11-3

11.2.2.1 World population external exposures at ground level


At ground level, the dominant component of the cosmic-ray field is muons with energies mostly between
1 and 20 GeV. These contribute about 80 % of the absorbed dose rate in free air from the directly ionizing
radiation. The remainder dose comes from electrons produced by the muons or present in the
electromagnetic cascade. As altitude increases, the electrons become more important contributors to the
dose rate.
According to the UNSCEAR 1988 Report [88U] the world population dose rate from directly ionizing
radiation is 31 nGy h1 at sea level. The dose rate is to be considered as averages over the 11-year solar
cycle, weighted by the fraction of 54 % of world population living in the northern and southern
hemisphere at latitudes below 30. Since mostly muons are involved, a radiation weighting factor of unity
is appropriate yielding the same values for the effective dose rate, i.e. 31 nSv h1.
The world average effective dose rate at sea level from neutrons of isotropic incidence obtained by
applying a neutron fluence energy distribution equally to 720 nSv h1 per neutron cm2 s1 is 5.5 nSv h1.
For both the ionizing and neutron components, there is a substantial altitude effect (see Fig. 11.1
[93U] and Fig. 11.2 [93U]). For the directly ionizing and photon component the population-weighted
average dose rate is 1.25 times that at sea level, for neutrons 2.5 times. Consequently the world effective
dose rate from exposures outdoors is 39 nSv h1 for the directly ionizing and photon component, and
14 nSv h1 for the neutron component.
Assuming a shielding effect of buildings of 20 % (shielding factor 0.8) and an indoor occupancy of
80 % of time for both cosmic radiation charged particles, photons and neutrons the world average
effective dose rate from the directly ionizing and photon component of cosmic rays is about 25 nSv h1 or
219 Sv a1, the corresponding average values for the neutron component are 9 nSv h1 or 78 Sv a1. The
total world average external annual effective dose is thus 297 Sv a1.
10 4

2.0
La Paz

Equivalent dose rate [mSv /a]

Absorbed dose rate [nGy h 1 ]

Solar minimum
Solar maximum

10 3

10 2 Ionizing component

10

1.5

0.5

Neutron component

1
10-1

10

Quito

1.0

Mexico City
Munich
New York

10 2

Altitude [km]
Fig. 11.1. Absorbed dose rate in air at 50 geomagnetic
latitude from the ionizing and neutron components of
cosmic rays as a function of altitude; [93U].

1
2
3
Hight above sealevel [10 3 m]

Fig. 11.2. Equivalent dose rate of selected populations


at various hights above sea level; data taken from [93U].

11.2.2.2 Exposures by cosmic radiations at aircraft altitudes


Aircraft passengers and crew are subject to cosmic radiation exposure rates partly much higher than the
rates at ground level, depending on the particular path taken through the atmosphere in terms of altitude
above sea level (see Fig. 11.1) and geomagnetic latitude (see. Fig. 11.3 [00U]).

11-4

11 Exposures from natural radiation sources

[Ref. p. 11-12

12

Effective dose rate [nSv h 1]

10
8
6
4

Measurements
Birattari et al.
Nakamura et al.
Fit to measurements

10

20

30 40 50 60 70
Geomagnetic latitude [deg]

80

100

Fig. 11.3. Geomagnetic latitude variation in effective dose


rate from cosmic ray neutrons at sea level; data taken from
[00U].

For altitudes of 9 - 12 km (commercial subsonic aircraft) at temperate latitudes, the effective dose
rates are in the range of 5 - 8 Sv h1, such that for a transatlantic flight of 6 hours from Europe to North
America the route dose would be 30 - 50 Sv. At equatorial latitudes, the dose rates are lower and in the
range of 2 - 4 Sv h1, such that for a 10 hours flight from Europe to South Africa the route dose would
be 20 - 40 Sv.
For crew members the average annual flight duration, i.e. the time between leaving the terminal before
take-off and returning after landing multiplied by the annual number of flights, is assumed to be 500
hours (300 - 900 h). For occasional flyers an average of 10 hours (3 - 50 h) and for frequent flyers
(business flyers or couriers) of 100 hours (50 - 1 200 h) is assumed annually.
A small portion of passengers and flight crews travel at higher altitudes of about 18 km on supersonic
transports. Effective dose rates of 10 - 20 Sv h1 are normally found with possible significant dose
contributions from solar particle events.
11.2.2.3 Internal exposures by cosmogenic radionuclides
The interactions of cosmic-ray particles in the atmosphere by high energy spallation interactions produce
a number of radionuclides, including 3H, 7Be, 14C and 22Na at a global inventory of 1275, 413, 12750, and
0.44 PBq, respectively. Only for these elements, which are of metabolic importance in the human body,
doses are worth mentioning. UNSCEAR [93U] previously assessed the annual effective doses from these
cosmogenic radionuclides to be 12 Sv (14C), 0.15 Sv (22Na), 0.03 Sv (7Be), and 0.01 Sv (3H),
respectively, or in total about 10 Sv.

11.3 Terrestrial radiation


Naturally occuring radionuclides of terrestrial origin primordial radionuclides are present in various
degrees in all media of the environment, including the human body itself. Only those radionuclides with
half-lives comparable to the age of the earth, and their decay products, exist in significant quantities in
these materials.
Irradiation of the human body from external sources is mainly by -radiation from radionuclides in the
238
U and 232Th series and from 40K. These radionuclides are also present in the human body from
ingestion and inhalation, and irradiate the various organs with - and -particles, as well as -rays. Some
other terrestrial radionuclides, including those of the 235U series, 87Rb, 138La, 147Sm, and 176Lu, exist in
nature but at such low levels that their contributions to the dose in humans are small.

Ref. p. 11-12]

11 Exposures from natural radiation sources

11-5

11.3.1 External exposures


11.3.1.1 Outdoors
External exposure outdoors arise from terrestrial radionuclides present at trace levels in all soils
depending on the types of rock from which the soils originate. Gamma-spectrometric measurements
indicate that the three components of the external radiation field, i.e. from the -emitting radionuclides in
the 238U and 232Th series and 40K, make approximately equal contributions to the externally incident
-radiation dose to individuals in typical situations both outdoors and indoors.
The activity concentration of 40K in soils is an order of magnitude higher than that of 238U or 232Th
series. UNSCEAR [00U] suggested median values for 40K, 238U, and 232Th series of 400, 35, and 30 Bq
kg1, and population-weighted values of 420, 33, and 45 Bq kg1. Based on corresponding dose
coefficients of 0.0417, 0.462, and 0.604 nGy h1 per Bq kg1 the total median and population weighted
absorbed dose rates in air were calculated to be 51 and 60 nGy h1, respectively. The lowest values of the
absorbed dose rate in air outdoors are in Cyprus, Iceland, Egypt, the Netherlands, Brunei, and the United
Kingdom, all less than 40 nGy h1, and the higher values are in Australia, Malaysia, and Portugal, all
greater than 80 nGy h1.
In addition to variations from place to place, the ambient background -dose rate in air at any specific
location is not constant in time. It is subject to considerable fluctuation, in particular from the removal of
222
Rn progeny in air by rainfall, soil moisture and snow cover.
There are small areas of markedly high absorbed dose rates in air throughout the world that are
associated with thorium-bearing and uranium-bearing minerals in soil, such as monazite sand deposits of
high levels of thorium as in Guarapari in Brazil (beaches, population size: 73 000; from 90 - 90 000
nGy h1), Yangiang in China (population size: 80 000; <370 nGy h1 average), the states of Kerala and
Madras in India (costal areas, population size: 100 000; 200 - 4 000 nGy h1), and Ramsar and Mahallat
in Iran (population size: 2 000; 70 - 17 000 nGy h1), in the latter areas caused by 226Ra deposited from
waters flowing from hot springs [00U].
In summary the population weighted absorbed dose rate in air outdoors from terrestrial -radiation is
60 nGy h1.
11.3.1.2 Indoors
Indoor exposure to -rays is mainly determined by the materials of construction and their surrounding
configuration indoors, and inherently greater than outdoor exposure if earth materials have been used.
When the duration of occupancy is taken into account, indoor exposure becomes even more significant.
From surveys of absorbed dose rates in air inside dwellings the population-weighted average of the
absorbed dose rate proved to be 84 nGy h1 with national averages ranging from 20 - 200 nGy h1 [00U].
The lowest values are in New Zealand, Iceland and the United States, all below 40 nGy h1, which
probably reflects the preponderance of wood-frame houses. The higher values (95 - 115 nGy h1) are in
Hungary, Malaysia, China, Albania, Portugal, Australia, Italy, Spain, Sweden, and Iran, which must
reflect wide use of stone or masonry materials in buildings.
11.3.1.3 Effective dose from external exposures
To estimate the annual effective doses, account must be taken of the conversion coefficient from absorbed
dose in air to effective dose, and the outdoor and indoor occupancy factors. The averages of the numerical
values of these parameters vary with the age of the population and the climate at the location considered.
In the UNSCEAR 1993 Report [93U], the Committee used 0.7 Sv Gy1 for the conversion coefficient
from absorbed dose in air to the effective dose received by adults, and 0.8 for the indoor occupancy
factor, i.e. the fraction of time spent indoors and outdoors is 0.8 and 0.2, respectively.

11-6

11 Exposures from natural radiation sources

[Ref. p. 11-12

From the data summarized in Sections 11.3.1.1 and 11.3.1.2 (outdoor population weighted absorbed
dose rate in air: 60 nGy h1; indoors: 84 nGy h1) the worldwide average of the effective dose rate is
55 nGy h1, and of the annual effective dose 486 Sv (outdoors: 74 Sv; indoors: 412 Sv). The latter is
for induvidual countries generally within the 300 - 600 Sv range. For children and infants the values are
about 10 % and 30 % higher depending on the conversion coefficient from absorbed dose in air to
effective dose.

11.3.2 Internal exposures


Internal exposures arise from the intake of terrestrial radionuclides by inhalation and ingestion. Doses by
inhalation result from the presence in air of dust particles containing radionuclides of the 238U and 232Th
decay chains.
The dominant component of inhalation exposure is the short-lived decay products of radon, which is
considered separately in Section 11.3.2.2.
Doses by ingestion are mainly due to 40K and to 238U and 232Th series radionuclides present in foods
and drinking water.
The dose rate from 40K can be determined directly from external measurements in vivo of its
concentration in the human body. The dose rate from uranium- and thorium-series radionuclides in the
body is estimated either from measured activity concentrations after chemical analyses of tissues or from
results of analyses of radionuclide contents of foods and drinking water, along with bioassay data and the
knowledge of the metabolic and biokinetic behaviour of the radionuclides.
11.3.2.1 Radionuclides other than radon
Intake by inhalation of natural radionuclides other than radon and its short-lived decay products makes
only a minor contribution to internal exposure. They cover long-lived radon decay products due to
disintegration of 222Rn in air and radionuclides of the 238U and 232Th series present in air because of
resuspended soil particles [00U]: 210Pb (500 Bq m3), 210Po (50 Bq m3), 238U (1 Bq m3), 235U
(0.05 Bq m3), 232Th and 230Th (each 0.5 Bq m3), 228Th (1 Bq m3), 228Ra and 226Ra (each 1 Bq m3).
That means the highest concentration to be for 210Pb, and those for the others to be lower by factors of 10,
500, 1 000, and 10 000.
Ingestion intake of natural radionuclides depends on the consumption rates of food and drinking water
and on the radionuclide concentrations. Based on reference consumption profiles, reference water balance
information and reference values for concentrations of uranium- and thorium-series radionuclides in
foods and drinking water UNSCEAR [00U] derived the follwing reference values for annual intakes:
210
Po (58 Bq), 210Pb (30 Bq), 228Ra (15 Bq), 226Ra (22 Bq), 238U (5.7 Bq), 230Th (3.0 Bq), 228Th (3.0 Bq),
232
Th (1.7 Bq), 235U (0.2 Bq).
The age-weighted (age distribution: infants 0.05, children 0.3, adults 0.65) committed annual effective
dose from inhalation of uranium- and thorium-series radionuclides in air is 5.8 Sv, that from ingestion is
roughly 25 times higher, i.e. 140 Sv. The annual committed effective dose from the reference values of
uranium- and thorium-series radionuclides in tissues evaluated in the UNSCEAR Report of 1988 [88U]
and adjusted to revised tissue weighting factors [93U] is 130 Sv in close agreement with the estimate of
110 Sv derived for adults from the dietary consumption by adults of reference concentrations of
radionuclides in foods and water.
Potassium is more or less uniformly distributed in the human body following intake in foods, and its
concentration in the body is under homeostatic control. The annual equivalent dose in tissues from 40K
and hence the annual effective dose is 165 and 185 Sv for adults and children, respectively, i.e. 170 Sv
weighted for age.

Ref. p. 11-12]

11 Exposures from natural radiation sources

11-7

The total committed annual effective dose from inhalation and ingestion of terrestrial radionuclides
weighted for age is 316 Sv of which 170 Sv is from 40K and 146 Sv is from the long-lived
radionuclides in the uranium and thorium series.
11.3.2.2 Radon and decay products
11.3.2.2.1 Sources, health risks and exposure-to-dose conversion
Inhalation of radon and its short-lived decay products in the atmosphere are the most important
contributors to human exposure from natural sources. 222Rn and 220Rn are the gaseous radioactive
products of the decay of the radium isotopes 226Ra and 224Ra, which are present in all terrestrial materials.
Some of the atoms of these radon isotopes are released from the solid matrix of the material by recoil
when radium decays, and escape from the mineral grain into the pore space. Radon atoms entering the
pore space are then transported by diffusion and advection through this space until they in turn decay or
are released into the atmosphere outdoors or indoors, i.e. into buildings (see Fig. 11.4 and 11.5 [01B]).

Long-lived
decay products

214
84 Po

214
83 Bi

218
84 Po

214
82 Pb

222
86 Rn

Diffusion

Convection
Diffusion

Aerosols

Rain trops

Sedimentation
Convection

Precipitation

Surface of the earth


Earth cracks
238
92 U

234
90 Th

234
91 Pa

234
92 U

230
90 Th

226
88 Ra

15%
222
86 Rn

85%

Decay products
in geological formations

Fig. 11.4. Formation of 222Rn by


decay of 226Ra in uranium-bearing
minerals, release of radon atoms from
mineral grain into pore space and
partial transportation by diffusion into
the atmosphere outdoors or indoors;
redrawn from [01B].

Inhalation of the short-lived decay products of 222Rn, and to a lesser extent of the decay products of
Rn (thoron), and their subsequent deposition along the walls of the various airways of the bronchial
tree provide the main pathway for radiation exposure of the lungs, predominantly by -particles. From
miners studies it is known that the -particle irradiation of the secretory and basal cells of the upper
airways is responsible for the lung cancer risk. Thus, the damage to these critical target cells of the
respiratory tract depends in a sensitive manner on the source/target geometry.
The dose that is relevant to the amount of lung-cancer risk depends critically on those environmental
factors that affect the probability of the radon decay products to be deposited near the critical target cells
after inhalation, i.e. the fraction of radon decay products which is attached to aerosols, the size
distribution of the aerosols, and the unattached fraction of radon decay products, as well as the radon
activity concentration, the equilibrium between radon and its decay products, the subjects inhalation rate,
and the time of exposure.
220

11-8

11 Exposures from natural radiation sources

e.g.15 Bq /m 3

[Ref. p. 11-12

e.g.50 Bq /m 3

e.g.120 Bq /m 3

e.g.2000 Bq /m

e.g.300 Bq /m 3

Fig. 11.5. Indoor diffusion of


Rn and its distribution in a building; redrawn from [01B].
222

Absorbed doses to the critical cells and effective doses are determined by applying exposure-to-dose
conversion factors. For 222Rn the range of these values derived from epidemiological studies and physical
dosimetry varies from 6 to 15 nSv (Bq h m3)1. As in its 1993 Report [93U] UNSCEAR applies in its
2000 Report [00U] the dose conversion factor of 9 nSv (Bq h m3)1. Since there are no epidemiological
data for lung cancer risk following 220Rn exposure from which to derive a conversion convention for
thoron decay products the value of 40 nSv (Bq h m3)1 derived from the ICRP Human Respiratory Tract
Model [94I] (see Chapter 7) was used for the estimation of equilibrium equivalent thoron doses for indoor
and outdoor exposures.
11.3.2.2.2 Air concentrations outdoors and indoors
Recent results of radon measurements outdoors tend to confirm the estimates of typical outdoor 222Rn and
220
Rn concentrations made in the UNSCEAR 1993 Report [93U] of 10 Bq m3 for each radon isotope
(range: for 222Rn 1 to >100 Bq m3; for 220Rn much smaller due to the short half-live).
The equilibrium factor, defined as the ratio of the actual potential -energy concentration to that, if all
the decay products in each series were in equilibrium with the parent radon, is suggested for radon in the
outdoor environment to be 0.6 for 222Rn, and 0.01 for 220Rn, respectively [00U].
For radon indoor concentrations the corresponding representative data are [00U]:
222
Rn activity concentration 40 Bq m3, equilibrium factor 0.4;
220
Rn activity concentration, 10 Bq m3, equilibrium factor 0.03.
11.3.2.2.3 Effective doses
For the above worldwide arithmetic outdoor and indoor radon gas concentrations, representative
equilibrium factors for the actual potential -energy concentration for outdoor and indoor occupancy
factors of 0.2 and 0.8 corresponding to annually 1760 and 7000 h, and the dose conversion factors of 9
and 40 nSv (Bq h m3)1 for 222Rn and 220Rn, respectively, the following annual effective doses are
derived [00U]:
222

Rn

Outdoors:
Indoors:

10 Bq m3 0.6 1 760 h 9 nSv (Bq h m3)1 = 95 Sv


40 Bq m3 0.4 7 000 h 9 nSv (Bq h m3)1 = 1 000 Sv

Ref. p. 11-12]
220

11 Exposures from natural radiation sources

11-9

Rn

Outdoors:
Indoors:

10 Bq m3 0.01 1 760 h 40 nSv (Bq h m3)1 = 7 Sv


10 Bq m3 0.03 7 000 h 40 nSv (Bq h m3)1 = 84 Sv

For completeness, the contribution to the annual effective dose from dissolution of the radon gases in
blood with distribution throughout the body is [00U]:
222

Rn

Outdoors:
Indoors:
220

3 Sv
48 Sv

Rn

Outdoors:
Indoors:

2 Sv
8 Sv

The total global annual average of the effective dose from inhalation of 222Rn and its decay products
present in air, from dissolution of radon gas in blood and ingestion of radon gas with tap water (2 Sv)
[93U] is 1148 Sv with fractions of about 95 % from inhalation outdoors and indoors and 5 % from
dissolved radon gas in blood and from ingestion of tap water. The annual effective dose from 220Rn is
101 Sv with fractions of about 90 % from inhalation and 10 % from thoron dissolution in blood.
These estimates of the global averages of the annual effective doses for radon only define the normal
radon and thoron exposures. One may expect to find many large populations around the world in the
order of 106 individuals, whose average exposures differ from the above global averages by a factor of
more than 2, and up to a factor of more than 10 for many smaller populations in the order of 104
individuals.

11.4 Enhanced exposures form industrial activities


There are numerous circumstances in which materials containing natural radionuclides are recovered,
processed and used, causing extra or enhanced population exposures. These exposures are those arising
from the mineral processing industries and from fossil fuel combustion by emission of radionuclides by
fly ash to air and water, and subsequent eventual intake by humans. Landfills after dredging or wastes
disposed on land may also provide pathways of exposure.
Main industries are:

Phosphate processing
Metal ore processing
Uranium mining
Fossil fuels for electric power production
Oil and gas extraction.

Estimated maximum exposures are greatest for phosphoric acid production and for the mineral-sandsprocessing industries. Although effective dose rates of the order of 100 Sv a1 could be received by a
few local residents, levels of the annual per caput effective doses of 1 - 10 Sv would be more common
[00U]. These exposure rates constitute a negligible component of the total annual effective doses from all
natural sources of radiation.

11-10

11 Exposures from natural radiation sources

[Ref. p. 11-12

11.5 Worldwide average exposure from natural and man-made sources


Worldwide average annual exposure by adding the various components described in Sections 11.2 to 11.4
proves to be about 2400 Sv effective dose. The sources of exposure and the single values of the
worldwide annual effective doses are summarized in Table 11.1 in Sv and in % of the total effective
dose, respectively. It should be stated that this average annual effective dose does not pertain to any one
individual, since there are wide distributions of exposures from each source, and the exposures combine
in various ways at each location, depending on the specific concentrations of radionuclides in the
environment and in the human body, the latitude and altitude of the location and many other factors such
as living habits.
Table 11.1. Average worldwide exposure to natural radiation sources
Source of exposure
Worldwide average
of the annual effective dose

Cosmic radiation
Directly ionizing and photon component
Neutron component
Cosmogenic radionuclides
Total exposure from cosmic
and cosmogenic sources
External terrestrial radiation
Outdoors
Indoors
Total external terrestrial exposure
Internal exposure: Inhalation
Uranium and thorium series
222
Rn + daughters
220
Rn + daughters
Total inhalation exposure
Internal exposure : Ingestion
40
K
Uranium and thorium series
Total ingestion exposure
Internal exposure: from blood
222
Rn, 220Rn + daughters
Enhanced exposures:
from industrial activities
Total (rounded)

[Sv a1]

[% of total]

219
78
10
307

9.1
3.2
0.4
12.7

300 - 2000

74
412
486

3.1
17.1
20.2

300 - 600

6
1148
101
1255

0.2
47.5
4.2
51.9

200 - 10000

170
140
310

7.1
5.8
12.9

200 - 800

45

1.9

10 - 400 (?)

<10
2400

<0.4
100.0

1 - 10
1000 - 10000

Typical range
[Sv a1]

The normal ranges of exposure to the various components of natural radiation are also indicated in
Table 11.1. This accounts for common variations in exposures, but excludes those individuals at extreme
ends of the distributions. On this basis, the worldwide average annual exposure to natural radiation
sources of 2400 Sv being the present estimate of the central value would generally be expected to be in
the range of 1000 - 10000 Sv. About 15 % of the worldwide average exposure is due to cosmic and
cosmogenic sources, about 20 % to external terrestrial exposure, in the order of 10 % is from ingestion of
natural radionuclides and about half of the total annual effective dose is due to the inhalation of both
radon isotopes 222Rn and 220Rn together with their radioactive decay products.

Ref. p. 11-12]

11 Exposures from natural radiation sources

11-11

The present (year 2000) worldwide total annual per caput effective dose from man-made sources is
about 410 Sv (400 Sv from diagnostic medical examinations, 5 Sv from atmospheric nuclear weapons
testing, 0.2 Sv from nuclear power production, and 2 Sv from the Tschernobyl nuclear reactor accident
[00U]), i.e. only about 15 % of the dose from natural sources. The largest contribution to exposures of
individuals worldwide is from medical diagnostic procedures (about 98 %), only 0.05 % from nuclear
power production.
There are a number of occupations, in which workers are exposed to enhanced natural and man-made
sources of radiation, i.e. to doses that are directly due to the work. Enhanced natural sources are air travel
(crew), mining (other than coal), coal mining, mineral processing, and radon at above ground work
places. Man-made sources are the nuclear fuel cycle (including uranium mining), industrial uses of
radiation, military defence activities, medical uses of radiation, education, and veterinary. The present
(year 2000) average annual effective dose from occupational exposure to enhanced natural sources is
1800 Sv, due to man-made sources is 600 Sv [00U], i.e. about 75 % and 25 % , respectively, of the
worldwide average annual per caput effective dose from natural background of 2400 Sv.

11-12

11 Exposures from natural radiation sources

11.6 References
87N
88U
93U
94I
96B
00U
01B

Nakamura, T., Y. Uwamino, T. Ohkubo: Altitude variation of cosmic-ray neutrons; Health


Physics 53(5) (1987) 509-517.
United Nations. Sources, Effects and Risks of Ionizing Radiation. United Nations Scientific
Committee on the Effects of Atomic Radiation, 1988 Report to the General Assembly, with
Annexes. United Nations, New York 1988.
United Nations. Sources and Effects of Ionizing Radiation. United Nations Scientific Committee
on the Effects of Atomic Radiation, 1993 Report to the General Assembly, with Annexes. United
Nations, New York 1993.
International Commission on Radiological Protection. Human Respiratory Tract Model for
Radiological Protection. Annals of the ICRP 24 (1 - 3). ICRP Publication 66. Pergamon Press,
Oxford, 1994.
Birattari, C., B. Moy, T. Rancati et al.: Neutron measurements at some environmental monitoring
stations; Internal Report. CERN, TIS-RP/IR/96-13 (1996).
United Nations. Sources and Effects of Ionizing Radiation. United Nations Scientific Committee
on the Effects of Atomic Radiation, 2000 Report to the General Assembly, with Annexes. United
Nations, New York 2000.
Bundesumweltministerium, Bundesamt fr Strahlenschutz: Radon-Handbuch Deutschland 2001,
BMU/BfS.

S-ar putea să vă placă și