Sunteți pe pagina 1din 42

Evolving Approaches

to Improve Outcomes
and Minimize Toxicities
in Radiation Therapy
San Francisco, Calif., USA, November 4, 2001

Guest Editor
Gillian M. Thomas, Toronto, Canada

24 figures and 17 tables, 1999

Basel 폷 Freiburg 폷 Paris 폷 London 폷 New York 폷


Bangalore 폷 Bangkok 폷 Singapore 폷 Tokyo 폷 Sydney
Supported by an educational grant from Ortho Biotech Products L.P.

S. Karger Drug Dosage All rights reserved.


Medical and Scientific Publishers The authors and the publisher have exerted every effort to en- No part of this publication may be translated into other
sure that drug selection and dosage set forth in this text are in languages, reproduced or utilized in any form or by any means,
Basel 폷 Freiburg 폷 Paris 폷 London
accord with current recommendations and practice at the time electronic or mechanical, including photocopying, recording,
New York 폷 Bangalore 폷 Bangkok of publication. However, in view of ongoing research, changes microcopying, or by any information storage and retrieval
Singapore 폷 Tokyo 폷 Sydney in government regulations, and the constant flow of informa- system, without permission in writing from the publisher or, in
tion relating to drug therapy and drug reactions, the reader is the case of photocopying, direct payment of a specified fee to
urged to check the package insert for each drug for any change the Copyright Clearance Center (see ‘General Information’).
in indications and dosage and for added warnings and precau-
tions. This is particularly important when the recommended © Copyright 2002 by S. Karger AG,
agent is a new and/or infrequently employed drug. P.O. Box, CH–4009 Basel (Switzerland)
Printed in Switzerland on acid-free paper by
Reinhardt Druck, Basel
ISBN 3–8055–7528–9

Fax + 41 61 306 12 34
E-Mail karger@karger.ch
www.karger.com
Vol. 63, Suppl. 2, 2002

Contents

1 Foreword
Thomas, G.M. (Toronto)

2 Radioprotectants: Current Status and New Directions


Grdina, D.J.; Murley, J.S.; Kataoka, Y. (Chicago, Ill.)

11 Prevalence of Anemia in Cancer Patients Undergoing Radiotherapy:


Prognostic Significance and Treatment
Harrison, L.B.; Shasha, D.; Homel, P. (New York, N.Y.)

19 Raising Hemoglobin: An Opportunity for Increasing Survival?


Thomas, G.M. (Toronto)

29 New Chemotherapeutic Agents: Update of Major Chemoradiation Trials


in Solid Tumors
Curran, W.J. (Philadelphia, Pa.)

© 2002 S. Karger AG, Basel

Fax + 41 61 306 12 34 Access to full text and tables of contents,


E-Mail karger@karger.ch including tentative ones for forthcoming issues:
www.karger.com www.karger.com/ocl_issues
Oncology 2002;63(suppl 2):1
DOI: 10.1159/00067144

Foreword

A variety of techniques are now available to radiation study of patients with head and neck cancers. Other
oncologists to optimize treatment of cancers, including potential applications of amifostine are reviewed in the
altered fractionation schedules, enhanced image guidance, article by Dr. Grdina and colleagues, along with recent
intensity modulation allowing radiation dose escalation advances in the development of newer cytoprotectants to
and improved brachytherapy techniques. In recent years, reduce the acute and chronic toxicities associated with
there has been increasing interest in the concurrent or high-dose treatment strategies and aggressive combined
sequential use of chemotherapeutic agents with radiosensi- modality protocols.
tizing ability to enhance the effectiveness of radiotherapy. The occurrence of anemia in cancer patients is an often
These agents include cisplatin, 5-fluorouracil, taxanes, to- overlooked complicating factor that is associated with
potecan, gemcitabine, vinorelbine, and tirapazamine. In poorer outcome possibly by decreasing the response to
certain malignancies (e.g., non-small-cell lung cancer, head radiotherapy, presumably via lowering the oxygen-carry-
and neck cancers, esophageal cancer and cervical cancer), ing capacity of the blood and thus exacerbating intratu-
concurrent chemotherapy and radiotherapy protocols have moral hypoxia. In addition, anemia has an adverse effect
resulted in better tumor control and/or patient survival on the quality of life of cancer patients, as evidenced by
than with radiotherapy alone. The review by Dr. Curran in the increased fatigue that has been associated with low
this supplement provides an update of recent clinical trials hemoglobin levels. Studies in various types of cancers
in this area, and emphasizes that while much has been have indicated that a high proportion of patients are
achieved in the quest for new combined modality regimens anemic prior to or during radiotherapy, and that low
capable of improving the outcomes for patients with can- hemoglobin levels are associated with poor clinical out-
cer, important questions concerning the selection of pa- comes with radiotherapy. As emphasized in other articles
tients, and the optimal dosages and timing of sequential in this supplement, these findings underline the impor-
therapies remain to be answered in future studies. tance of early detection and treatment of anemia in cancer
Other evolving approaches to optimizing radiotherapy patients. Administration of epoetin alfa to correct anemia
include the use of radioprotectants to reduce radio- has been reported to enhance locoregional response rates
therapy-induced toxicity without affecting its antitumor to chemoradiation therapy in patients with certain types
efficacy, cytotoxic agents such as mitomycin C to specifi- of cancers (e.g., oropharyngeal squamous cell carcinomas)
cally target hypoxic tumor cells, and strategies to counter and to improve quality of life. Whether epoetin alfa thera-
anemia such as treatment with epoetin alfa (recombinant py will also increase long-term survival is currently being
human erythropoietin). It is postulated that anemia in investigated. Other ongoing studies are investigating
cancer patients may result in a poor treatment outcome whether epoetin alfa may also be effective in protecting
because of an increased resistance to radiation or chemo- against radiotherapy-induced neurotoxicity.
therapy. Radioprotectants currently under investigation The challenge for the future is to utilize our present
include amifostine (WR-1065), which has been shown in knowledge to optimize the management of cancer pa-
experimental studies to prevent both radiation-induced tients undergoing radiotherapy or combined modality
cell death and radiation-induced mutagenesis. Moreover, protocols with the objective of improving both the out-
this agent reduced the incidence of early and late radio- come of treatment and quality of life.
therapy-induced xerostomia in a multicenter clinical Gillian M. Thomas

© 2002 S. Karger AG, Basel Gillian M. Thomas


ABC 0030–2414/02/0636–0001$18.50/0 Radiation Oncology, Obstetrics & Gynecology
Fax + 41 61 306 12 34 University of Toronto
E-Mail karger@karger.ch Accessible online at: Toronto-Sunnybrook Regional Cancer Centre
www.karger.com www.karger.com/ocl Toronto, Onta. (Canada)
Oncology 2002;63(suppl 2):2–10
DOI: 10.1159/000067146

Radioprotectants: Current Status and


New Directions
David J. Grdina Jeffrey S. Murley Yasushi Kataoka
Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, Ill., USA

Key Words (MnSOD). In addition, the use of epoetin alfa, alone or in


Cytoprotection W Radiotherapy W Radiation-induced combination with cytoprotectants (e.g., amifostine), to
toxicity W Mutagenesis W Amifostine W Thiol compounds treat radiation-induced anemia is also being investi-
gated. The objective of developing newer cytoprotective
therapies is to improve the therapeutic ratio by reducing
Abstract the acute and chronic toxicities associated with more
The ability to prevent radiotherapy-induced toxicity with- intensive and more effective anticancer therapies.
out affecting antitumor efficacy has the potential to Copyright © 2002 S. Karger AG, Basel

enhance the therapeutic benefit for cancer patients with-


out increasing their risk of serious adverse effects.
Among the currently available cytoprotective agents ca- Introduction
pable of protecting normal tissue against damage
caused by either chemo- or radiotherapy, only amifos- Radiotherapy is toxic not only toward cancer cells but
tine has been shown in clinical trials to reduce radiation- also to healthy cells, particularly those with a high rate of
induced toxicity. Most notably, it reduces the incidence proliferation, which may result in serious adverse effects
of xerostomia, which is a clinically significant long-term for patients. The risk of cell toxicity is increased with the
toxicity arising in patients undergoing irradiation of head application of more intensive radiotherapy techniques
and neck cancers. In vitro studies with the active metabo- intended to increase tumor cell kill. Radiation-induced
lite of amifostine (WR-1065) have shown it to prevent adverse effects commonly include mucositis and/or der-
both radiation-induced cell death and radiation-induced matitis, and are usually managed symptomatically as they
mutagenesis. The potential of this agent to prevent sec- manifest. However, preventing these complications is
ondary tumors, as well as other radiation-induced toxici- clearly more desirable, and various approaches to reduc-
ties is now the focus of ongoing research. Among other ing radiation-induced toxicities while maintaining antitu-
novel approaches to radioprotection being explored are mor efficacy have been investigated. These include al-
methods to increase levels of the antioxidant mito- tered radiation dose fractionation, the use of physical
chondrial enzyme manganese superoxide dismutase shielding or intensity modulated radiation therapy to

© 2002 S. Karger AG, Basel David J. Grdina


ABC 0030–2414/02/0636–0002$18.50/0 Department of Radiation and Cellular Oncology
Fax + 41 61 306 12 34 University of Chicago Medical Center, MC 1105
E-Mail karger@karger.ch Accessible online at: 5841 S. Maryland Avenue, Chicago, IL 60637 (USA)
www.karger.com www.karger.com/ocl Tel. +1 773 702 5250, Fax +1 773 702 5940, E-Mail dgrdina@rover.uchicago.edu
reduce the volume of exposure, and pharmacologic ap-
proaches. The latter can be divided into radiosensitizers 100
which ideally differentially enhance the sensitivity of
tumors rather than normal tissue, and radioprotectants to
reduce the detrimental effects of radiation on normal tis-

or normal tissue damage (%)


Probability of tumor control
75
Tumor
sue while maintaining tumor sensitivity [1, 2]. This article
reviews the current status of radioprotectants in cancer Normal tissue

therapy and provides an insight into some of the new 50


directions that research in this area is taking.
Typical response curves illustrating the probability of
tumor control and normal tissue damage at varying radia- 25
tion doses are shown in figure 1. The objective of radio-
protection is to shift the response curve for normal tissue
as far as possible to the right to achieve the highest proba- 0
A B C
bility of tumor control with the least amount of damage to Radiation dose
normal tissue. The ideal radioprotectant is one that pro-
tects normal tissue while preserving antitumor effective-
ness, and is itself without moderate or severe toxicity. Fig. 1. Tumor and normal tissue response curves to radiotherapy,
illustrating the probability of tumor control and normal tissue dam-
age at varying radiation doses (reproduced with permission from
Hall [12]).
Pharmacologic Strategies for Cytoprotection
of Normal Cells

In recent years, a number of cytoprotective agents


capable of protecting normal tissue against damage WR-1065. This metabolite is then oxidized to the disul-
caused by either chemo- or radiotherapy have been devel- fide form WR-33278 [4–6]. Numerous preclinical studies
oped. As a result of studies implicating toxic metabolites have shown that amifostine protects normal cells against
of chemotherapeutic agents and/or the generation of high- the adverse effects of both radiation and chemotherapeut-
ly reactive species or free radicals in the etiology of DNA ic agents (e.g., alkylating agents, platinum compounds,
damage [3–5], a number of different strategies have been anthracyclines and taxanes) without attenuating their cy-
proposed for cytoprotection of normal cells, including: totoxic effects on large solid tumors. This selective protec-
E preventing the generation of toxic metabolites of che- tion is due, in part, to the more efficient conversion and
motherapeutic agents; uptake of the active metabolite WR-1065 in normal tissue
E enhancing the elimination of toxic metabolites of che- in comparison with neoplastic tissue, as a result of the
motherapeutic agents; higher alkaline phosphatase activity, greater vasculariza-
E neutralizing DNA adduct-forming metabolites; tion, and higher pH of normal tissue [4–6].
E detoxifying free radicals. Following intravenous administration, amifostine is
A number of compounds have been investigated with rapidly and extensively taken up by normal tissue. Ani-
the objective of providing site-specific protection for nor- mal studies have indicated that maximal concentrations
mal tissues without compromising the antitumor efficacy of the active metabolite WR-1065 occur 5–15 min after
of chemotherapeutic agents and/or radiotherapy, includ- administration [7]. Uptake of WR-1065 in normal tissue
ing amifostine (WR-2721), dexrazoxane, mesna, gluta- is not uniform, and appears to be greatest in the kidney,
thione, and N-acetylcysteine. Among these, amifostine, salivary glands, intestinal mucosa, liver and lung [6].
dexrazoxane and mesna have FDA approval for use in Once inside the cell, WR-1065 protects against chemo-
cytoprotection. therapy- and radiotherapy-induced DNA damage by (1)
binding to and neutralizing the reactive species of organo-
Amifostine platinum and alkylating agents, thus preventing forma-
Amifostine (WR-2721) is a nucleophilic sulfur prodrug tion of adducts with DNA, and (2) scavenging free radi-
that is dephosphorylated in vivo by membrane-bound cals [5–7].
alkaline phosphatase to the active, free thiol metabolite

Prevention of Radiation-Induced Toxicity Oncology 2002;63(suppl 2):2–10 3


Table 1. Net charges of thiol compounds with putative cytoprotec- metabolism of dexrazoxane and/or differences in its uptake
tive activity between normal cardiac cells and tumor cells [5, 9–11].
Currently, dexrazoxane has FDA approval to reduce
Compound Net charge
the incidence and severity of cardiomyopathy associated
WR-33278 (disulfide metabolite of amifostine) +4 with doxorubicin administration in women with meta-
WR-1065 (free thiol metabolite of amifostine) +2 static breast cancer who have received cumulative doses
Cystamine +2 1300 mg/m2.
Cysteamine +1
Captopril 0
Dithiothreitol (DTT) 0
Mesna
2-Mercaptoethanol (2-ME) 0 Mesna (sodium 2-mercaptoethane sulfonate) was de-
N-Acetyl-L-cysteine (L-NAC) –1 veloped as a specific chemoprotectant against the toxicity
N-Acetyl-D-cysteine (D-NAC) –1 of acrolein, a urotoxic metabolite of oxazaphosphorine-
Mesna –1
based alkylating agents (e.g., ifosfamide and cyclophos-
Glutathione, reduced (GSH) –1
Glutathione, oxidized (GSSG) –2 phamide), which produces hemorrhagic cystitis following
its excretion into the urinary bladder. Following intrave-
nous administration, mesna is converted into an inactive
disulfide form in the blood and is then metabolized back
to mesna in the urinary tract where its free sulfhydryl
The efficacy of amifostine in protecting cancer patients groups bind to and inactivate acrolein, forming a stable,
against radiotherapy-induced toxicity is discussed below. non-toxic thioether that is rapidly excreted in the urine.
Currently, amifostine has FDA approval to reduce the Mesna also inhibits the further formation of acrolein in
incidence of xerostomia in patients undergoing radiation the bladder. Because its activity is restricted to the urinary
treatment for head and neck cancers. It is also approved to tract, the systemic activity and non-urologic toxicity of
reduce cumulative renal toxicity associated with cisplatin oxazaphosphorine drugs are not affected [4, 5].
treatment in patients with ovarian cancer or non-small- Currently, mesna has FDA approval for the prophylax-
cell lung cancer. is of ifosfamide-induced hemorrhagic cystitis.

Dexrazoxane
Dexrazoxane (ICRF-187) is a cyclic derivative of the Relationship Between the Net Charge of Thiol
metal-chelating agent ethylenediamine-tetraacetic acid Compounds and Their Ability to Protect
(EDTA) that provides protection against the cardiotoxici- Against Radiation-Induced DNA Damage
ty of anthracycline-based chemotherapeutic agents, such
as doxorubicin. Although the risk of cardiotoxicity ap- The mechanism by which radiation induces DNA
pears to be reduced with newer formulations, such as damage is slightly different to that of chemotherapeutic
peglyated liposomal doxorubicin [8], cardiotoxicity is a agents. Radiation-induced damage is introduced into a
well-recognized, serious, treatment-limiting adverse ef- genome by either a direct action, where the energy is
fect of these compounds. It occurs via the generation of deposited directly on the genome, or indirectly via the for-
reactive oxygen species, which are highly toxic to cardiac mation of free radicals which are responsible for the resul-
tissues, by the stable complexes formed between anthra- tant cell killing, mutagenesis, transformation, and carci-
cycline drugs and iron [9]. The cardioprotective effect of nogenesis. The latter mechanism, which accounts for
dexrazoxane is believed to result from its intracellular about 75% of radiation-induced DNA damage by pho-
metabolism to a ring-opened hydrolysis product (ICRF- tons, can be abrogated with free radical scavengers
198), which is a strong chelator of free and bound intracel- present in the local microenvironment at the time the free
lular iron in the myocardium. As a consequence, the radicals are formed. However, in the case of direct dam-
amount of iron available to form complexes with anthra- age, there are no known radioprotectants as this process
cyclines is reduced and formation of the reactive oxygen occurs too rapidly to be prevented by a pharmacologic
species is blocked. Importantly, the protective effect of agent [12].
dexrazoxane against the cardiotoxicity of anthracycline Studies performed several years ago by Fahey et al.
drugs occurs without affecting their antitumor activity. have shown that the net charge of thiol compounds with
This may be due, in part, to differences in the intracellular putative cytoprotective activity (table 1) markedly in-

4 Oncology 2002;63(suppl 2):2–10 Grdina/Murley/Kataoka


1.0
a a
1.0

Surviving fraction of cells


Surviving fraction of cells

0.1
0.1

60
Co γ - radiation
60
Co g-radiation WR - 1065 after 60
Co γ - radiation
WR-1065 before Co γ-radiation
60
0.01
0.01 100 b

HPRT mutants per 106 survivors


100 b
80
mutants per 106 survivors

80

60
60
HPRT

40 40

20 20

0 0
0 2 4 6 8 10 12 0 2 4 6 8 10 12
60
Co g dose (Gy) 60
Co γ dose (Gy)

Fig. 2. Response to varying doses of 60Co Á-radiation of V79 Chinese Fig. 3. Response to varying doses of 60Co Á-radiation of V79 Chinese
hamster lung fibroblast cells in the absence or presence of WR-1065 hamster lung fibroblast cells exposed immediately after irradiation to
4 mmol/l added to cell cultures 30 min before irradiation and allowed WR-1065 4 mmol/l added to cell cultures and allowed to remain for
to remain until 3 h after irradiation. a Surviving fraction of cells. 3 h. a Surviving fraction of cells. b Mutation induction at the HPRT
b Mutation induction at the HPRT (hypoxanthine-guanine phospho- (hypoxanthine-guanine phosphoribosyl transferase) locus among sur-
ribosyl transferase) locus among surviving cells that were grown in a viving cells (assessed as described in fig. 2). The broken lines repre-
non-selective medium for 6 days and then exposed to 6-thioguanine sent the radiation-only curves and are presented for comparison.
5 Ìg/ml in ·-MEM-10 medium (containing hypoxanthine, aminop- Bars represent the standard errors of the mean of two or more repli-
terin and thymidine) for 7 days and stained with 0.5% methylene cate experiments (reproduced with permission from Grdina et al.
blue. Bars indicate the standard errors of the mean of two or more [15]).
replicate experiments (reproduced with permission from Grdina et
al. [15]).

fluences the degree of protection that they provide against Protective Effect of WR-1065 (Amifostine Metabolite)
the DNA-damaging effects of radiation [13, 14]. Because Studies in our institution using V79 Chinese hamster
WR-1065 has a net charge of +2, it will be attracted to lung fibroblast cells have shown that WR-1065 in a con-
DNA (which is negatively charged) and is therefore more centration of 4 mmol/l protects against radiation-induced
likely to exert a protective effect against radiation- cell death when added to cell cultures 30 min before var-
induced damage than a compound with a net charge of 0 ious doses of 60Co Á-radiation, but not when it is added
or a negative net charge. Evidence in support of this immediately after irradiation (fig. 2a, 3a). This treatment-
hypothesis has come from studies with WR-1065, capto- schedule dependence in the protective effect of WR-1065
pril, and N-acetylcysteine in Chinese hamster lung fibro- is to be expected if it is acting as a free radical scavenger,
blast and ovary cells. since protection could only be expected to occur when the
compound is present during irradiation [15].

Prevention of Radiation-Induced Toxicity Oncology 2002;63(suppl 2):2–10 5


10 0
10 0
60
Co γ-radiation 60
Co -radiation
Surviving fraction of cells

Surviving fraction of cells


10 -1

10 -1

Radiation only
10 -2 Radiation +
4 mmol/l N-acetylcysteine
Without captopril Radiation +
0.04 mmol/l N-acetylcysteine
With 1 mmol/l captopril

10 -2 10 - 3
0 200 400 600 800 1,000 0 200 400 600 800 1,000
60
Co γ dose (cGy) 60
Co γ dose (cGy)

Fig. 4. Surviving fractions of Chinese hamster ovary (CHO)-AA8 Fig. 5. Surviving fractions of Chinese hamster ovary (CHO)-AA8
cells exposed to varying doses of 60Co Á-radiation in the absence or cells exposed to varying doses of 60Co Á-radiation in the absence or
presence of captopril 1 mmol/l added to the cell cultures 30 min prior presence of N-acetylcysteine 0.04 mmol/l and 4 mmol/l added to the
to irradiation (Grdina DJ, unpubl. data). cell cultures 30 min prior to irradiation (Grdina DJ, unpubl. data).

As well as increasing the surviving fraction of cells of a protective effect against radiation-induced cell killing
when administered before irradiation, WR-1065 has also with either drug (fig. 4, 5) [Grdina DJ, unpubl. data].
been shown to reduce the degree of radiation-induced Thus, key factors governing the radioprotective effica-
mutagenesis in V79 Chinese hamster lung fibroblast cells cy of a drug acting as free radical scavenger are: (1) an
(expressed as the HPRT mutant frequency per 106 survi- ability to concentrate within the nucleus or microenviron-
vors exposed to 6-thioguanine 5 Ìg/ml) (fig. 2b). In con- ment of DNA (dependent on its net charge), and (2) the
trast to the treatment-schedule dependence for the protec- presence of the protectant at the radiation target at the
tive effect against cell killing, the antimutagenic effect of time it is irradiated (important for prevention of cell
WR-1065 is also observed when it is administered after death). No clinical advantage is achieved if the protector
irradiation (fig. 3b), indicating that its post-irradiation does not differentially protect normal tissues compared to
action can effectively alter mutation induction in surviv- tumor.
ing cells [16].

Lack of Protective Effect of Captopril and Dose-Response Considerations with


N-Acetylcysteine Amifostine: Prevention of Cell Death vs
Because the net charges of captopril and N-acetylcys- Prevention of Mutagenesis
teine are 0 and –1, respectively, these thiol compounds
would not be expected to concentrate within the nega- The protection factor achievable with a radioprotec-
tively charged nucleus or the microenvironment of DNA tant is defined as the ratio of surviving cell fraction for
to the same extent as those with positive net charges. treated cells as compared with untreated cells following
Studies using Chinese hamster ovary (CHO)-AA8 cells radiation exposure. The clinical potential of a putative
exposed to various doses of 60Co Á-radiation in the radioprotectant depends on the tolerability of the drug at
absence or presence of captopril 1 mmol/l and N-acetyl- a dosage required to achieve a particular protection fac-
cysteine 0.04 mmol/l or 4 mmol/l have shown no evidence tor. In studies conducted at our institution using CHO-

6 Oncology 2002;63(suppl 2):2–10 Grdina/Murley/Kataoka


AA8 cells, the protection factor for cell survival with the
amifostine metabolite WR-1065 (i.e., the ratio of cell sur- 8
viving fractions for WR-1065-treated versus untreated
cells) was determined at various concentrations of WR-
1065 added to the incubation medium 30 minutes prior
to exposure to a radiation dose of 750 cGy from a 60Co 6

Protection factor (cell survival)


Á-ray source. As shown in figure 6, the protection factor
fell sharply from 16 to around 3 as the concentration of
WR-1065 was decreased from 4 to 1 mmol/l, and then
declined further to essentially no protection at lower con- 4

centrations of 0.01–0.1 mmol/l. In contrast, the protec-


tion against mutagenesis (expressed as the HPRT mutant
frequency per 106 survivors exposed to 6-thioguanine 5
Ìg/ml) remained largely constant over the same WR-1065 2

concentration range (0.01–4 mmol/l). This suggests that


the mechanism by which WR-1065 provides protection
against mutagenesis differs from that for protection
0
against cell killing, and that the antimutagenic effect 0.01 1 4
0.1
can be achieved at lower concentrations (as low as WR-1065 concentration (mmol/l)
0.01 mmol/l) [16].
Viewed in relation to therapeutic use of amifostine, the
concentrations of WR-1065 achieved with dosages of am- Fig. 6. Protection factor for survival of Chinese hamster ovary
ifostine used clinically are in the range 1.5–3.85 mmol/l (CHO)-AA8 cells (i.e., the ratio of cell surviving fractions for WR-
1065-treated to untreated cells) at varying concentrations of WR-
[17], which suggests that the degree of cytoprotection pro-
1065 added to the incubation medium 30 min prior to exposure to a
vided at antimutagenic dosages are insufficient to in- radiation dose of 750 cGy from a 60Co Á-ray source. All plot points
crease survival of either normal or neoplastic cells. How- are the average of three separate experiments and the bars represent
ever, its effect in reducing the risk of radiation-induced the standard errors of the mean (reproduced with permission from
mutagenesis, carcinogenesis, and secondary tumors is of Grdina et al. [16]).
considerable interest and this area is now an important
focus for ongoing research into the protective effects of
amifostine, particularly in view of increasing evidence
that the risk of secondary tumors is increased as cancer mas [18]. Patients in this multicenter study (n = 303)
therapies become more effective and, coincidentally, received radiotherapy in a dose of 1.8 to 2.0 Gy/day for 30
more damaging to normal tissues. to 35 fractions (total dose 50–70 Gy), approximately half
of whom (n = 153) were randomized to receive amifostine
(200 mg/m2 intravenously over 3 min) 15 to 30 min be-
Clinical Studies of Amifostine as a fore irradiation and half to receive radiotherapy alone
Radioprotectant (n = 150). As shown in table 2, amifostine significantly
reduced the incidence of both early xerostomia within the
Clinical trials of the radioprotective effect of amifos- first 90 days (as well as the cumulative radiotherapy dose
tine have been undertaken in patients receiving radiother- required to cause this adverse effect) and late xerostomia
apy for head and neck, pelvic, and thoracic cancers [6]. at 1 year after initiation of radiotherapy, although it did
Thus far, most studies have involved relatively small not significantly reduce the incidence of acute mucositis.
numbers of patients but have generally demonstrated sig- Patients who received amifostine were also found to pro-
nificant reductions in the incidence of radiation-induced duce more saliva than those treated with radiotherapy
local toxicities. In the largest trial conducted to date, the alone (median saliva production 0.26 vs 0.10 g; p = 0.04).
efficacy of amifostine in ameliorating the adverse effects When overall survival data for the two groups of patients
of radiotherapy and its influence on the clinical effective- were compared, there was a slight advantage for those
ness of radiotherapy were evaluated in patients with pre- receiving amifostine (fig. 7), but the difference was not
viously untreated head and neck squamous cell carcino- statistically significant. Nor was there any significant dif-

Prevention of Radiation-Induced Toxicity Oncology 2002;63(suppl 2):2–10 7


100
(124)
90
(104)
80 (119)

70 (98)

Percent survival
60

50 Total No.
Fig. 7. Percentages of survivors over a peri-
Events of patients
od of 27 months among patients with pre- 40 Amifostine + radiotherapy 34 153
viously untreated head and neck squamous
30 Radiotherapy alone 45 180
cell carcinomas who were randomized to
receive either amifostine (200 mg/m2 i.v. 20
Log-rank: p = 0.184
over 3 min) 15–30 min before radiotherapy Hazard ratio: 1.351 (95% Cl 0.865 - 2.109)
doses of 1.8–2 Gy/day for 30 to 35 fractions 10
(total 54–70 Gy) or similar doses of radio- 0
therapy alone. The numbers of patients at
0 3 6 9 12 15 18 21 24 27
risk at 12 and 18 months are indicated in Months
parentheses (reproduced with permission
from Brizel et al. [18]).

Table 2. Incidence of acute and late


xerostomia 6grade 2 (RTOG acute/late Complication Amifostine plus Radiotherapy p value
morbidity scoring criteria) in patients with radiotherapy alone
head and neck squamous cell carcinomas (n = 153) (n = 150)
who received radiotherapy with or without
amifostine (200 mg/m2 i.v. 15–30 min Acute xerostomiaa
prior to irradiation) [18] Incidence (% of patients) 51% 78% ! 0.0001
Cumulative radiotherapy
dose to onset 60 Gy 42 Gy ! 0.0001
Late xerostomiab
Incidence (% of patients) 34% 57% ! 0.002

RTOG = Radiation Therapy Oncology Group.


a Within 90 days of initiation of radiotherapy.
b At 1 year after initiation of radiotherapy.

ference between the two groups in locoregional tumor lung cancer and reduction of toxicities associated with
control rates. Thus, amifostine significantly reduced acute doxorubicin- and paclitaxel-containing regimens, high-
and chronic xerostomia in these patients without com- dose chemotherapies, and multimodality chemotherapy
promising the antitumor effectiveness of radiotherapy and radiotherapy for a variety of solid tumors. Possible
[18]. prevention of secondary tumors (see above), is also being
explored. In addition, the observation that amifostine
may stimulate bone marrow progenitor cells has led to
Potential Future Applications of Amifostine studies of its use as a potential treatment for patients with
myelodysplastic syndrome. As yet, however, clinical data
Other potential roles for amifostine that are being are limited and its value in this setting remains to be clari-
explored include reducing renal toxicity associated with fied [6].
cisplatin treatment in ovarian cancer and non-small-cell

8 Oncology 2002;63(suppl 2):2–10 Grdina/Murley/Kataoka


New Advances in Cytoprotection metabolites WR-1065 and WR-33278, N-acetylcys-
teine, mesna, captopril, oltipraz, and dithiothreitol
A number of newer potential radioprotectants are cur- [21–23]. In human microvascular endothelial cells,
rently undergoing preclinical research, including: (1) The exposure to WR-1065 0.04 mmol/l for 30 min has been
amifostine analog S-[2-(3-methylaminopropyl) aminoe- shown to cause an increase in MnSOD gene expression
thyl] phosphorothioate acid, which is orally bioavailable that begins about 12 h after exposure to WR-1065,
and less toxic than amifostine; (2) thiolamine compounds peaks at 16 to 18 h, and ends after about 22 h [21].
with thioglycoside-protecting groups; (3) covalent conju- E The use of epoetin alfa (recombinant human erythro-
gates of thioamines and antioxidant vitamins, and (4) sel- poietin; r-HuEPO) alone or in combination with cyto-
enazolidine prodrugs. protectants (e.g., amifostine) to treat radiation-induced
In addition, other approaches to radioprotection are anemia. The interaction of amifostine with epoetin
also being explored. These include altering endogenous alfa may produce a synergy in gene activation/expres-
levels of antioxidant enzymes (specifically the mitochon- sion (e.g., of the c-myb gene thereby leading to an
drial enzyme manganese superoxide dismutase [MnSOD] increase in hematopoietic progenitor cells), as well as
which protects against oxidative stress induced by various an increase in myeloproliferation and a reduction of
agents including irradiation), and enhancement of eryth- genomic instability [24–26].
ropoiesis to treat the anemia that commonly occurs dur- The objective of developing newer cytoprotective ther-
ing irradiation (see review by Harrison in this supple- apies is to be able to reduce the acute and cumulative toxi-
ment). Novel approaches that are currently being investi- cities associated with more intensive and more effective
gated include: therapeutic anticancer regimens now being introduced
E The use of MnSOD plasmid/liposome complex gene into clinical practice, whether delivered as radiotherapy,
therapy to protect against radiation-induced esophagi- chemotherapy, or combined modality regimens. The
tis. Improved tolerance of the esophageal epithelium to merging of these technologies will, it is hoped, enhance
fractionated radiation has recently been demonstrated the therapeutic benefit for cancer patients without in-
with this approach in a mouse model [19, 20]. creasing their risk of serious adverse effects, and thus
E The use of nonprotein thiol-containing compounds to improving both their quality and duration of life.
activate MnSOD gene expression, e.g., the amifostine

References

1 Curran WJ: Radiation-induced toxicities: the 8 Safra T, Muggia F, Jeffers S, Tsao-Wei DD, 12 Hall EJ: Radiobiology for the radiologist, ed 5.
role of radioprotectants. Semin Radiat Oncol Groshen S, Lyass O, Henderson R, Berry G, Philadelphia, Lippincott Williams & Wilkins,
1998;8(4 suppl 1):2–4. Gabizon A: Pegylated liposomal doxorubicin 2000.
2 Brizel DM: Future directions in toxicity pre- (doxil): reduced clinical cardiotoxicity in pa- 13 Zheng S, Newton GL, Gonick G, Fahey RC,
vention. Semin Radiat Oncol 1998;8(4 suppl tients reaching or exceeding cumulative doses Ward JF: Radioprotection of DNA by thiols:
1):17–20. of 500 mg/m2. Ann Oncol 2000;11:1029– relationship between the net charge on a thiol
3 Schuchter LM: Current role of protective 1033. and its ability to protect DNA. Radiat Res
agents in cancer treatment. Oncology (Hun- 9 Wiseman LR, Spencer CM: Dexrazoxane: a 1988;114:11–27.
tingt) 1997;11:505–512, 515–518. review of its use as a cardioprotective agent in 14 Zheng S, Newton GL, Ward JF, Fahey RC:
4 Hoekman K, van der Vijgh WJF, Vermorken patients receiving anthracycline-based chemo- Aerobic radioprotection of pBR322 by thiols:
JB: Clinical and preclinical modulation of che- therapy. Drugs 1998;56:385–403. effect of thiol net charge upon scavenging of
motherapy-induced toxicity in patients with 10 Hasinoff BB: The iron(III) and copper(II) com- hydroxyl radicals and repair of DNA radicals.
cancer. Drugs 1999;57:133–155. plexes of adriamycin promote the hydrolysis of Radiat Res 1992;130:183–193.
5 Links M, Lewis C: Chemoprotectants: A re- the cardioprotective agent ICRF-187 ((+)-1,2- 15 Grdina DJ, Nagy B, Hill CK, Wells RL, Perai-
view of their clinical pharmacology and thera- bis(3,5-dioxopiperazinyl-1-yl)propane). Agents no C: The radioprotector WR1065 reduces ra-
peutic efficacy. Drugs 1999;57:293–308. Actions 1990;29:374–381. diation-induced mutations at the hypoxan-
6 Culy CR, Spencer CM: Amifostine: an update 11 Koning J, Palmer P, Franks CR, Mulder DE, thine-guanine phosphoribosyl transferase locus
on its clinical status as a cytoprotectant in Speyer JL, Green MD, Hellmann K: Cardio- in V79 cells. Carcinogenesis 1985;6:929–931.
patients with cancer receiving chemotherapy or xane – ICRF-187 towards anticancer drug 16 Grdina DJ, Shigematsu N, Dale P, Newton
radiotherapy and its potential therapeutic ap- specificity through selective toxicity reduction. GL, Aguilera JA, Fahey RC: Thiol and disul-
plication in myelodysplastic syndrome. Drugs Cancer Treat Rev 1991;18:1–19. fide metabolites of the radiation protector and
2001;61:641–684. potential chemopreventive agent WR-2721 are
7 Schuchter LM: Guidelines for the administra- linked to both its anti-cytotoxic and anti-muta-
tion of amifostine. Semin Oncol 1996;23(4 genic mechanisms of action. Carcinogenesis
suppl 8):40–43. 1995;16:767–774.

Prevention of Radiation-Induced Toxicity Oncology 2002;63(suppl 2):2–10 9


17 Shaw LM, Bonner HS, Schuchter L, Schiller J, 20 Epperly MW, Kagan VE, Sikora CA, Gretton 24 List AF: Use of amifostine in hematologic ma-
Lieberman R: Pharmacokinetics of amifostine: JE, Defilippi SJ, Bar-Sagi D, Greenberger JS: lignancies, myelodysplastic syndrome, and
effects of dose and method of administration. Manganese superoxide dismutase-plasmid/li- acute leukemia. Semin Oncol 1999;26(2 suppl
Semin Oncol 1999;26(2 suppl 7):34–36. posome (MnSOD-PL) administration protects 7):61–65.
18 Brizel DM, Wasserman TH, Henke M, Strnad mice from esophagitis associated with fraction- 25 Jongen-Lavrencic M, Peeters HR, Vreugdenhil
V, Rudat V, Monnier A, Eschwege F, Zhang J, ated radiation. Int J Cancer 2001;96:221–231. G, Swaak AJ: Interaction of inflammatory cy-
Russell L, Oster W, Sauer R: Phase III random- 21 Murley JS, Kataoka Y, Hallahan DE, Roberts tokines and erythropoietin in iron metabolism
ized trial of amifostine as a radioprotector in JC, Grdina DJ: Activation of NFkappaB and and erythropoiesis in anaemia of chronic dis-
head and neck cancer. J Clin Oncol 2000;18: MnSOD gene expression by free radical scav- ease. Clin Rheumatol 1995;14:519–525.
3339–3345. engers in human microvascular endothelial 26 Zhu J, Heyworth CM, Glasow A, Huang QH,
19 Epperly MW, Gretton JA, DeFilippi SJ, Green- cells. Free Radic Biol Med 2001;30:1426– Petrie K, Lanotte M, Benoit G, Gallagher R,
berger JS, Sikora CA, Liggitt D, Koe G: Modu- 1439. Waxman S, Enver T, Zelent A: Lineage restric-
lation of radiation-induced cytokine elevation 22 Das KC, Lewis-Molock Y, White CW: Activa- tion of the RARalpha gene expression in my-
associated with esophagitis and esophageal tion of NF-kappa B and elevation of MnSOD eloid differentiation. Blood 2001;98:2563–
stricture by manganese superoxide dismutase- gene expression by thiol-reducing agents in 2567.
plasmid/liposome (SOD2-PL) gene therapy. lung adenocarcinoma (A549) cells. Am J Physi-
Radiat Res 2001;155:2–14. ol 1995;269(5 Pt 1):L588–L602.
23 Antras-Ferry J, Maheo K, Chevanne M, Dubos
MP, Morel F, Guillouzo A, Cillard P, Cillard J:
Oltipraz stimulates the transcription of the
manganese superoxide dismutase gene in rat
hepatocytes. Carcinogenesis 1997;18:2113–
2117.

10 Oncology 2002;63(suppl 2):2–10 Grdina/Murley/Kataoka


Oncology 2002;63(suppl 2):11–18
DOI: 10.1159/000067147

Prevalence of Anemia in Cancer Patients


Undergoing Radiotherapy:
Prognostic Significance and Treatment
Louis B. Harrison a Daniel Shasha a Peter Homel b
a Department of Radiation Oncology, Beth Israel Medical Center/St Luke’s-Roosevelt Hospital Center;
b Department of Grants and Research, Beth Israel Medical Center, New York, N.Y., USA

Key Words hemoglobin in patients with certain malignancies. Radia-


Radiotherapy W Anemia W Hypoxia W Radiation-induced tion oncologists need to be aware of the possibility of
toxicity W Epoetin alfa W Quality of life anemia in cancer patients undergoing radiotherapy so
that timely intervention can be instituted whenever ane-
Abstract mia is diagnosed.
As the antitumor activity of radiation is mediated via its Copyright © 2002 S. Karger AG, Basel

interaction with oxygen to form labile free radicals, the


intratumoral oxygen level has an important influence on
the ability of radiation therapy to kill malignant cells. By Introduction
decreasing the oxygen-carrying capacity of the blood,
anemia may result in tumor hypoxia and may have a The objective of radiotherapy in cancer treatment is to
negative influence on the outcome of radiotherapy for maximize locoregional tumor control and patient surviv-
various malignancies, even for small tumors not normal- al. As the antitumor activity of radiation is known to be
ly assumed to be hypoxic. In addition, anemia also has a mediated via its interaction with oxygen to form labile
negative effect on the quality of life of cancer patients, as free radicals, the intratumoral oxygen level has an impor-
evidenced by worsening fatigue. As a high proportion tant influence on the number of free radicals produced
(about 50%) of cancer patients undergoing radiotherapy within a tumor and thus on the ability of radiation thera-
are anemic prior to or during treatment, strategies to cor- py to induce DNA damage in malignant cells. Conse-
rect anemia and/or the resultant tumor hypoxia are quently, the presence of acute or chronic anemia, which
increasingly being considered an important component may decrease the oxygen-carrying capacity of the blood
of treatment. In particular, epoetin alfa (recombinant and results in tumor hypoxia, lowers the propensity of
human erythropoietin), which has proved an effective radiotherapy to produce DNA damage and is an obstacle
and well-tolerated means of raising hemoglobin levels in to achieving maximal locoregional tumor control [1–3]. It
anemic patients receiving radiotherapy, potentially has been estimated that the dose of radiation required to
could reverse the negative prognostic influence of a low kill tumor cells under hypoxic conditions is 2 to 3 times

© 2002 S. Karger AG, Basel Louis B. Harrison, MD


ABC 0030–2414/02/0636–0011$18.50/0 Department of Radiation Oncology, Beth Israel Medical Center
Fax + 41 61 306 12 34 10 Union Square East, New York, NY 10003 (USA)
E-Mail karger@karger.ch Accessible online at: Tel. +1 212 844-8087, Fax +1 212 844-8086
www.karger.com www.karger.com/ocl E-Mail Lharrison@bethisraelny.org
patients with head and neck cancers, a pretreatment
1 Hypoxic
hemoglobin level !11.0 g/dl was found to be a stronger
Normoxic
Fraction of survivng cells

predictor of poor tumor oxygenation than other factors


such as tumor stage, tumor volume and smoking status
0.1
[4]. Even prostate tumors, which are generally not con-
sidered to be hypoxic, have been reported to be associat-
0.01 OER = 1,750 ⴜ 700
ed with significantly lower pO2 levels in comparison
= 2.5 with pathologically normal prostate tissue and muscles,
particularly in patients with more advanced (T2/T3)
0.001 prostatic tumors and in older individuals (662 years of
0 200 400 600 800 1,000 1,200 1,400 1,600 1,800 2,000
age) [5].
Radiation dose (cGy)

Effect on Locoregional Tumor Control and Survival


Recent studies have demonstrated an important rela-
Fig. 1. Enhancement of radiation resistance by hypoxia. The oxygen
enhancement ratio (OER) is the ratio of the radiation dose required tionship between anemia on locoregional tumor control
to kill a given fraction of malignant cells in a hypoxic environment in and patient survival, principally in head and neck can-
relation to that in a normoxic environment. cers. In patients with early stage glottic cancers, which are
amongst the smallest tumors treated by oncologists, a
clear relationship has been demonstrated between the pre-
treatment hemoglobin level and the hazard ratio for local
the dose required in a normoxic environment (fig. 1). relapse following radiotherapy (50 Gy in 20 fractions over
Hypoxia has also been found to produce mutations of the 4 weeks) during a median follow-up period of 6.8 years
p53 suppressor gene, which results in an increase in angio- [8]. Similarly, studies in patients with squamous cell car-
genesis and an increased tendency for the development of cinomas of the glottic larynx and head/neck who were
distant metastases. Thus, overcoming hypoxia may have treated with radiotherapy have noted significantly better
positive effects on not only locoregional tumor control but 2-year locoregional tumor control rates and 2-year surviv-
also on decreasing the risk of developing metastatic dis- al rates in those who presented with normal hemoglobin
ease [1, 3]. levels in comparison with those who presented with
Hypoxia is a common characteristic of solid tumors. below-normal hemoglobin levels (!13 g/dl) (table 1) [9,
Athough the number and size of their hypoxic regions var- 10]. In the patients with head/neck cancers, 5-year locore-
ies substantially [1], hypoxia may be present in even small gional control and survival rates were also significantly
tumors at an early stage of development. In the past, the better in those with normal hemoglobin levels (p ! 0.001
prevalence of anemia and tumor hypoxia in cancer pa- and p ! 0.01, respectively; table 1) [10].
tients receiving radiotherapy has been an underappre- Other studies have shown a relationship between the
ciated problem that has frequently led to undertreatment. post-radiotherapy hemoglobin level and the outcome of
This article reviews the prevalence of anemia in patients treatment. Among patients with squamous cell carcino-
undergoing radiotherapy, and emphasizes that effective mas of either the glottic or supraglottic regions who
reversal of anemia can be achieved. Until recently, little received primary radiotherapy in doses ranging from 60
attention has been paid to hemoglobin levels in cancer to 70 Gy over 6 to 7 weeks, disease-free survival rates
patients. were significantly better in those who had normal hemo-
globin levels (defined as 12–16 g/dl in women, 13.7–
18 g/dl in men) at day 35 of treatment in comparison with
Prognostic Significance of Anemia in Cancer those who had below-normal hemoglobin levels at this
Patients time (p = 0.0012 for glottic carcinoma; p = 0.05 for supra-
glottic carcinoma) (fig. 2) [11].
Relationships between low hemoglobin levels and in-
tratumoral hypoxia, and between intratumoral hypoxia Effect on Fatigue
and a less favorable prognosis in various cancers have In addition to locoregional tumor control and survival,
been identified in studies in which oxygen partial pres- other outcomes in patients undergoing radiotherapy may
sures (pO2) were measured in tumor tissue [4–7]. In also be influenced by the presence of anemia. Fatigue is

12 Oncology 2002;63(suppl 2):11–18 Harrison/Shasha/Homel


40 Not anemic 37%
Anemic

Percentage of patients with


30 29% 28%]

a fatigue rating >5


20

14%

10
4%

0
Pre-radiotherapy During radiotherapy Post-radiotherapy
Time relative to radiotherapy

Fig. 2. Disease-free survival among patients with squamous cell car- Fig. 3. Percentages of prostate cancer patients with fatigue ratings
cinoma of the glottic and supraglottic regions treated with primary greater than 5 (on a scale of 0–10) before, during and after radiother-
radiotherapy (60–70 Gy in 30–35 fractions over 6–7 weeks) in rela- apy in relation to whether they were anemic (Hb level ! 12 g/dl) or
tion to their hemoglobin (Hb) levels at day 35 of treatment. p Values not anemic at the time (Harrison LB, unpublished data). * Statistical-
indicate differences between disease-free survival in each group for ly significant versus non-anemic patients (p = 0.015).
patients with normal vs below normal Hb levels. Normal Hb values
were defined as 13.7–18 g/dl (8.5–11.0 mmol/l) for men and 12–
16 g/dl (7.5–10.0 mmol/l) for women (reproduced with permission
from van Acht et al. [11]).

Table 1. Influence of anemia on locoregional tumor control rates and survival rates in two studies in patients with
squamous cell carcinomas of the glottic larynx (n = 109) or head/neck region (n = 504) [9, 10]

Patient group Locoregional control rates, % Survival rates, %


2-year 5-year 2-year 5-year

Glottic squamous cell carcinomas [9]


Normal hemoglobin levels 95* NR 88** NR
Anemiaa 66 NR 46 NR
Head/neck squamous cell carcinomas [10]
Normal hemoglobin levels 52** 48** 51* 36*
Anemiab 34 32 37 22

a Hemoglobin ! 13 g/dl.
b Hemoglobin ! 13 g/dl (women) or ! 14.5 g/dl (men).
* p ! 0.01 vs anemic patients; ** p ! 0.001 vs anemic patients; NR = not reported.

one such outcome that has been strongly associated with in those who were anemic (hemoglobin !12 g/dl) than in
anemia [12–14]. In a group of patients with prostate can- those who were not anemic (fig. 3). This finding is of
cer treated with radiotherapy at our institution, the per- interest because fatigue is generally not considered a prob-
centage with a fatigue rating 15 (on a scale of 0–10) fol- lem in prostate cancer patients receiving radiotherapy
lowing radiotherapy was found to be significantly higher alone.

Prevalence and Treatment of Oncology 2002;63(suppl 2):11–18 13


Radiotherapy-Associated Anemia
100 Baseline
During radiotherapy
77% 79%

Patients with anemia (%)


80 75%

63%
55% 32%
60
Fig. 4. The prevalence of anemia (Hb ! 12 44% 45% 44%
g/dl) before and during radiotherapy in pa- 40 16%
tients with different types of cancer treated 26%
at the Department of Radiation Oncology,
20
Beth Israel Medical Center/St Luke’s-Roo- 9%
sevelt Hospital Center, New York between
December 1996 and June 1999. Baseline 0
Breast Colorectal Lung/ Prostate Uterine/ Head/
was defined as within 4 weeks prior to the cancer cancer bronchus cancer cervical neck
first radiation dose. During therapy was de- (n = 81) (n = 64) cancer (n = 90) cancer cancer
(n = 64) (n = 53) (n = 68)
fined as within 3 to 5 weeks of the first radia-
tion dose.

41% of patients were found to be anemic at baseline


2.0
]
(within 4 weeks prior to radiotherapy) and 54% were
1.8 ]
Mean decrease in hemoglobin

anemic within 3 to 5 weeks after receiving the first dose of


during radiotherapy (g/dl)

1.6 ]
radiation [15]. The prevalence of anemia was higher in
1.4
women than in men (54 vs 28% at baseline; 63 vs 43%
1.2 ]
during radiotherapy), and was higher in patients with cer-
1.0
tain types of cancer than others (fig. 4). In particular, high
0.8
0.6
prevalences of anemia were noted in patients with colo-
0.4
rectal, lung/bronchus and uterine/cervical cancers, and
0.2
increases in prevalence from baseline to end of therapy
0 were most notable for those with colorectal and lung/
Breast Colorectal Lung Prostate Cervical Head/ bronchus cancers (fig. 4). Among patients who experi-
cancer cancer cancer cancer cancer neck
(n = 71) (n = 48) (n = 101) (n = 78) (n = 48) cancer enced a drop in their hemoglobin level during radiothera-
(n = 86)
py, the mean decreases ranged from 0.75 g/dl for those
with breast cancer to 1.8 g/dl for those with head or neck
Fig. 5. Mean decreases in hemoglobin (Hb) levels during radiothera- cancers, and the decreases were statistically significant
py versus preradiotherapy in patients treated at the Department of (p ! 0.001) in all groups except those with breast and cer-
Radiation Oncology, Beth Israel Medical Center/St Luke’s-Roosevelt
vical cancer (fig. 5).
Hospital Center, New York between December 1996 and June 1999.
Data shown are for patients whose Hb levels decreased during treat- When the prevalence of anemia for each cancer type
ment. * Statistically significant difference versus baseline (p ! was stratified by the hemoglobin level measured at base-
0.001). line and the lowest level recorded during radiotherapy,
most patients in each group were found to have mild ane-
mia (hemoglobin levels 610 g/dl), which should be easily
correctable. These data, and the findings of studies re-
Prevalence of Anemia in Patients Undergoing viewed previously in this article indicating that the pres-
Radiotherapy for Various Cancers ence of anemia is associated with poorer treatment out-
comes, provide compelling evidence for employing strate-
Studies of patients presenting for radiotherapy at our gies to correct anemia and/or the resultant tumor hypoxia
institution between December 1996 and June 1999 (n = in cancer patients undergoing radiotherapy.
574) have revealed a high prevalence of anemia (hemoglo-
bin !12 g/dl) both before and during irradiation. Overall,

14 Oncology 2002;63(suppl 2):11–18 Harrison/Shasha/Homel


100 HBO4 Carbogen patients at 18 months (n = 36)
Air
Local relapse-free rate (%)

Carbogen patients at 3 years (n = 36)


80 [estimated probabilities]
Noncarbogen patients at 18 months (n = 36)
100
60 91% 91%

40 80 75% 75%
69%

Patients (%)
20 62% 62%
60 55%
50%
0
0 1 2 3 4 5 40
Years
20

Fig. 6. Local relapse-free survival over 5 years in patients with locally 0


advanced squamous cell carcinomas of the head or neck who were Local control Cause-specific survival Overall survival
randomized to treatment with either radiotherapy under hyperbaric
oxygen at 4 atmospheres (HBO4) delivered in two fractions of
11.5 Gy over 21 days (n = 23), or radiotherapy delivered in air in two Fig. 7. Influence of carbogen breathing on local control, cause-spe-
fractions of 12.65 Gy over 21 days (n = 25) (reproduced with permis- cific survival and overall survival in patients with advanced head or
sion from Haffty et al. [17]). neck cancers treated with a hyperfractionated chemoradiotherapy
regimen. Patients received either carboplatin 5 mg/m2 administered
45 min before radiotherapy (115 cGy) with carbogen breathed 4 min
prior to and during irradiation twice per day on 5 days a week for 7
weeks (n = 36), or the same chemoradiotherapy regimen without car-
Strategies to Correct Anemia and/or Tumor
bogen breathing (comparison group; n = 36). Data at 3 years for the
Hypoxia carbogen breathing group are estimated probabilities [18].

Strategies that have been proposed to correct anemia


and/or the resultant tumor hypoxia include the use of:
E Hypoxic cell sensitizers (e.g., cytotoxic agents) rate in comparison with the accelerated regimen alone (48
E Fluosol infusion vs 34% and 39 vs 28%, respectively), indicating that hyp-
E Carbogen breathing oxia can, in part, be overcome by mitomycin C adminis-
E Hyperbaric oxygen tration. Mitomycin C did not influence the local toxicity
E Blood transfusions of radiotherapy as neither the intensity nor the duration
E Epoetin alfa (recombinant human erythropoietin; of radiotherapy-induced mucositis was altered by its ad-
r-HuEPO). ministration [16].

Hypoxic Cell Sensitizers Hyperbaric Oxygen and Carbogen Breathing


In a study designed to evaluate the efficacy of the cyto- The use of hyperbaric oxygen to overcome tumor hyp-
toxic agent mitomycin C in sensitizing hypoxic tumor oxia has been reported to produce an improved response
cells to the effects of radiotherapy, patients with squa- to hypofractionated radiotherapy in a randomized trial in
mous cell carcinomas of the head or neck were treated patients with advanced squamous cell carcinoma of the
with either conventional fractionated radiotherapy head or neck. Patients who received radiotherapy under
(70 Gy/35 fractions/7 weeks) or continuous hyperfrac- hyperbaric oxygen at 4 atmospheres showed a higher 5-
tionated accelerated radiotherapy (55.3 Gy/17 consecu- year local relapse-free rate than those who received a simi-
tive days/33 fractions) with or without mitomycin C lar radiotherapy regimen delivered in air (29 vs 16%;
(20 mg/m2) given on day 5 of treatment [16]. Local tumor fig. 6). However, there were no significant differences
control and survival rates over a median follow-up period between the two groups in 5-year survival, distant metas-
of 148 months were similar with the two radiotherapy tasis, or second primary tumors [17].
regimens given alone; however, the addition of mitomy- Similarly, carbogen breathing has also been shown to
cin C to the accelerated regimen significantly reduced improve the results of chemoradiotherapy (carboplatin
both the local tumor control rate and the overall survival 5 mg/m2 given before radiation doses of 115 cGy twice

Prevalence and Treatment of Oncology 2002;63(suppl 2):11–18 15


Radiotherapy-Associated Anemia
Table 2. Influence of the pretreatment hemoglobin level and epoetin alfa on the outcome of therapy in patients
undergoing chemoradiation plus surgical treatment for squamous cell carcinomas of the oral cavity or oropharynx
[24]

Patient group Overall complete 2-year locoregional 2-year


response, %a tumor control, % survival, %

Group 1: patients with Hb 614.5 g/dl not treated


with epoetin alfa (n = 43) 65* 88** 81**
Group 2: patients with Hb ! 14.5 g/dl not treated
with epoetin alfa (n = 87) 17 72 60
Group 3: patients with Hb ! 14.5 g/dl treated
with epoetin alfab (n = 57) 61* 95* 88*

* p (0.001 compared with group 2; ** p ! 0.05 compared with group 2; Hb = hemoglobin; SC = subcutaneously.
a Complete responses were determined by histopathologic analysis of the en bloc resection of the primary tumor and
regional cervical lymphatics performed 5 to 6 weeks after the completion of chemoradiotherapy.
b Dosage: 10,000 IU/kg SC 3 to 6 times per week until week of surgery.

daily on 5 days per week for 7 weeks) in patients with therapy in our earlier study of the prevalence of anemia in
locally advanced head or neck cancer. Anemic patients patients with various malignancies (fig. 5).
also received either blood transfusions or epoetin alfa to The effects of epoetin alfa on the outcomes of therapy
correct the anemia. Patients who breathed carbogen have been studied in anemic patients (Hb !14.5 g/dl)
4 min before and during irradiation exhibited improved with squamous cell carcinomas of the oral cavity or oro-
local control, cause-specific survival, and overall survival pharynx [23, 24]. All patients in this study received a regi-
at 18 months in comparison with a similar number of men consisting of mitomycin C (15 mg/m2 on day 1), 5-
patients who received the same chemoradiotherapy regi- fluorouracil (750 mg/m2 on days 1–5) and radiotherapy
men without carbogen breathing (fig. 7). The high re- (50 Gy in 25 fractions during weeks 1–5), followed by dis-
sponse rates achieved in this study appeared to persist as section of the primary tumor bed and a neck dissection.
the estimated probabilities of local control, cause-specific Epoetin alfa (10,000 IU/kg subcutaneously 3 to 6 times
survival, and overall survival at 3 years in the carbogen per week until the week of surgery) was administered to a
breathing group were similar to the rates observed at 18 group of patients (n = 57) who had a pretreatment hemo-
months [18]. globin level !14.5 g/dl. The outcome in this group of
patients was compared with the outcomes in two other
Epoetin Alfa (Recombinant Human Erythropoietin; groups who did not receive epoetin alfa. One of these non-
r-HuEPO) epoetin alfa groups had a pretreatment hemoglobin level
The ability of epoetin alfa to correct anemia prior to !14.5 g/dl (n = 87) and the other had a pretreatment
and during radiotherapy has been evaluated in cancer hemoglobin level 614.5 g/dl (n = 43). The results are
patients to determine whether it produces clinically summarized in table 2. In the two groups of patients who
meaningful benefit. Studies in patients receiving radio- did not receive epoetin alfa, those with a low pretreatment
therapy for various malignancies have shown that the hemoglobin level (!14.5 g/dl) (group 2) exhibited signifi-
administration of epoetin alfa, with or without oral iron, cantly lower complete response rates, 2-year locoregional
is effective in increasing hemoglobin levels and is well tol- control rates, and 2-year survival rates than those who
erated [19–21]. A study in our institution in cancer had normal hemoglobin levels (614.5 g/dl) (group 1).
patients receiving a variety of different chemotherapy reg- However, in the patients with a low pretreatment hemo-
imens with concomitant or sequential radiotherapy has globin level who received epoetin alfa (group 3), the rates
shown that weekly epoetin alfa administration improved of complete response, 2-year locoregional control and 2-
the mean hemoglobin level by 1.8–3.4 g/dl [22] (fig. 5). year survival were equivalent to or higher than those in
Improvements of this magnitude are similar to or greater patients with normal pretreatment hemoglobin levels
than the reductions in hemoglobin noted during radio- (group 1) [24].

16 Oncology 2002;63(suppl 2):11–18 Harrison/Shasha/Homel


These findings suggest that epoetin alfa is an effective the results of studies in our institution in which visual
and well-tolerated means of achieving normal hemoglo- evoked potentials (VEPs) were measured in animals re-
bin levels in patients undergoing radiotherapy, and may ceiving radiotherapy in the presence and absence of r-
reverse the negative prognostic influence of a low pre- HuEPO. Pretreatment of animals with epoetin alfa signif-
treatment hemoglobin level. Improvements in quality-of- icantly prolonged VEPs as compared with those not
life parameters (linear analog scale assessment) have also receiving epoetin alfa, suggesting that it may protect
been noted with epoetin alfa therapy in groups of patients visual pathways against radiation-induced damage (A.
receiving a variety of different chemotherapy regimens Evans, unpublished data). If confirmed clinically, this
with concomitant or sequential radiotherapy [22]. finding may have substantial implications for the use of
radiotherapy in patients with malignancies of the head,
paranasal sinuses and ocular regions because it suggests
Potential Benefit of Epoetin Alfa in Reducing that epoetin alfa may provide biologic protection of the
Radiotherapy-Induced Neurotoxicity optic nerve against radiation-induced damage.

In addition to studies of the efficacy of epoetin alfa in


improving the clinical outcome of radiotherapy in pa- Conclusions
tients with low hemoglobin levels, its potential to reduce
radiation-induced neurotoxicity is also being investi- Anemia may result in tumor hypoxia by decreasing the
gated. Studies in experimental animals have revealed that oxygen-carrying capacity of the blood, resulting in radia-
endogenous erythropoietin (EPO) possesses other biologi- tion and, in some instances, chemotherapy resistance.
cal activities in addition to erythropoietic effects, and that Anemia is associated with a poorer prognosis in a variety
many cells besides erythroid progenitors express the of malignancies. It may be an important obstacle to
erythropoietin receptor, including brain cells. As in the achieving maximal locoregional tumor control and sur-
periphery, erythropoietin production is known to be in- vival with radiotherapy, even for small tumors not nor-
duced by hypoxia in the central nervous system (CNS), mally assumed to be hypoxic. In addition, anemia nega-
and it has been shown in animals to protect CNS neuronal tively affects the quality of life of cancer patients, as evi-
cells from ischemic injury [25]. A recent study found that denced by worsening fatigue. In view of the high preva-
erythropoietin receptors are abundantly expressed in cap- lence of anemia recorded in cancer patients receiving
illaries of the brain-periphery interface, suggesting that radiotherapy (about 50% at our institution), it is evident
this may provide a route for circulating erythropoietin to that measures to reverse anemia and tumor hypoxia
enter the brain [26]. In support of this hypothesis, a study should be considered an important component of treat-
in mice showed that systemic administration of epoetin ment for such patients. Indeed, a number of strategies,
alfa (5,000 IU/kg intraperitoneally) 24 h before or up to notably the administration of epoetin alfa, have been
6 h after controlled blunt trauma to the frontal cortex and found to attenuate the negative prognostic influence of a
then continued once daily for 4 additional days (5 doses low hemoglobin level in patients receiving radiotherapy
total) attenuated the resultant brain injury. Quantitative with or without chemotherapy.
analysis of the cavitary injury volume showed that the These findings indicate the need for radiation oncolog-
concussive injury in mice treated with epoetin alfa was ists to be aware of the possibility of anemia in cancer
significantly less than in those treated with saline. In addi- patients undergoing radiotherapy so that timely interven-
tion, epoetin alfa also ameliorated the damage caused by tion with strategies to improve the outcome of treatment
experimentally-induced focal ischemic stroke in rat can be instituted whenever anemia is diagnosed. In view
brains, reduced the severity of experimental autoimmune of the potential benefits of treating anemia, it is hoped
encephalitis in Lewis rats, and delayed and lessened sei- that this aspect of cancer management will receive more
zures induced in mice by the glutamate analog kainic attention in the future.
acid. These findings in different models of neurologic
injury suggest that epoetin alfa is able to cross the blood-
brain barrier and may provide protection against CNS
neurologic damage [26].
Further evidence in support of a protective effect of
erythropoietin against neurologic damage is provided by

Prevalence and Treatment of Oncology 2002;63(suppl 2):11–18 17


Radiotherapy-Associated Anemia
References

1 Shasha D: The negative impact of anemia on 10 Lee WR, Berkey B, Marcial V, Fu KK, Cooper 20 Dusenbery KE, McGuire WA, Holt PJ, Carson
radiotherapy and chemoradiation outcomes. JS, Vikram B, Coia LR, Rotman M, Ortiz H: LF, Fowler JM, Twiggs LB, Potish RA: Eryth-
Semin Hematol 2001;38(3 suppl 7):8–15. Anemia is associated with decreased survival ropoietin increases hemoglobin during radia-
2 Kumar P: Tumor hypoxia and anemia: impact and increased locoregional failure in patients tion therapy for cervical cancer. Int J Radiat
on the efficacy of radiation therapy. Semin with locally advanced head and neck carcino- Oncol Biol Phys 1994;29:1079–1084.
Hematol 2000;37(4 suppl 6):4–8. ma: a secondary analysis of RTOG 85-27. Int J 21 Sweeney PJ, Nicolae D, Ignacio L, Chen L,
3 Dunst J: Hemoglobin level and anemia in ra- Radiat Oncol Biol Phys 1998;42:1069–1075. Roach M 3rd, Wara W, Marcus KC, Vijayaku-
diation oncology: prognostic impact and thera- 11 van Acht MJ, Hermans J, Boks DE, Leer JW: mar S: Effect of subcutaneous recombinant hu-
peutic implications. Semin Oncol 2000;27(2 The prognostic value of hemoglobin and a de- man erythropoietin in cancer patients receiving
suppl 4):4–8, 16–17. crease in hemoglobin during radiotherapy in radiotherapy: final report of a randomised,
4 Becker A, Stadler P, Lavey RS, Hansgen G, laryngeal carcinoma. Radiother Oncol 1992; open-labelled, phase II trial. Br J Cancer 1998;
Kuhnt T, Lautenschlager C, Feldmann HJ, 23:229–235. 77:1996–2002.
Molls M, Dunst J: Severe anemia is associated 12 Groopman JE: Fatigue in cancer and HIV/ 22 Shasha D, George M, Harrison LB: Once-
with poor tumor oxygenation in head and neck AIDS. Oncology (Huntingt) 1998;12:335–344. weekly dosing of epoetin alfa increases hemo-
squamous cell carcinomas. Int J Radiat Oncol 13 Sabbatini P: Contribution of anemia to fatigue globin and improves quality of life in anemic
Biol Phys 2000;46:459–466. in the cancer patient. Oncology (Huntingt) cancer patients receiving radiation therapy
5 Movsas B, Chapman JD, Greenberg RE, Hor- 2000;14(11A):69–71. either concurrently or sequentially with chemo-
witz EM, Pinover WH, Hanlon AL, Stobbe C, 14 Sobrero A, Puglisi F, Guglielmi A, Belvedere therapy. Presented at the 42nd Annual Meeting
Hanks GE: Increasing levels of hypoxia in hu- O, Aprile G, Ramello M, Grossi F: Fatigue: a of the American Society of Hematology, San
man prostate carcinoma correlate significantly main component of anemia symptomatology. Francisco, CA, Dec 2000.
with increasing clinical stage and age: an Ep- Semin Oncol 2001;28(2 suppl 8):15–18. 23 Glaser CM, Millesi W, Kornek GV, Lang S,
pendorf pO2 study. Int J Radiat Oncol Biol 15 Harrison LB, Shasha D, Shiaova L, et al: Preva- Schüll B, Klug K, F, Wanschitz F, Lavey RS:
Phys 1999;45(3 suppl):202. lence of anemia in cancer patients undergoing Impact of hemoglobin (Hgb) level and use of
6 Brizel DM, Dodge RK, Clough RW, Dewhirst radiotherapy (abstract). Proc Am Soc Clin On- recombinant human erythropoietin (r-HuEPO)
MW: Oxygenation of head and neck cancer: col 2000;19:471a. on response to neoadjuvant chemoradiation
changes during radiotherapy and impact on 16 Dobrowsky WH, Naudé J, Widder J, Dobrow- therapy, tumor control, and survival in patients
treatment outcome. Radiother Oncol 1999;53: sky E: Continuous hyperfractionated acceler- with oral or oropharyngeal squamous cell carci-
113–117. ated radiotherapy and mitomycin C in head noma (SCCA). Int J Radiat Oncol Biol Phys
7 Höckel M, Vorndran B, Schlenger K, Bauss- and neck cancer. Int J Radiat Oncol Biol Phys 1999;45(3 suppl):149.
mann E, Knapstein PG: Tumor oxygenation: a 1999;45(3 suppl):148. 24 Glaser CM, Millesi W, Kornek GV, Lang S,
new predictive parameter in locally advanced 17 Haffty BG, Hurley R, Peters LJ: Radiation Schüll B, Watzinger F, Selzer E, Lavey RS:
cancer of the uterine cervix. Gynecol Oncol therapy with hyperbaric oxygen at 4 atmo- Impact of hemoglobin level and use of recom-
1993;51:141–149. spheres pressure in the management of squa- binant erythropoietin on efficacy of preopera-
8 Warde P, O’Sullivan B, Bristow RG, Panzarel- mous cell carcinoma of the head and neck: tive chemoradiation therapy for squamous cell
la T, Keane TJ, Gullane PJ, Witterick IP, results of a randomized clinical trial. Cancer J carcinoma of the oral cavity and oropharynx.
Payne D, Liu FF, McLean M, Waldron J, Cum- Sci Am 1999;5:341–347. Int J Radiat Oncol Biol Phys 2001;50:705–
mings BJ: T1/T2 glottic cancer managed by 18 Martinez A, Cabezon M, Fuentes C, Espiñeira 715.
external beam radiotherapy: the influence of M, Perez M, Serdio J, Artazkoz J, Gil J, Borque 25 Sakanaka M, Wen TC, Matsuda S, Masuda S,
pretreatment hemoglobin on local control. Int J C, Villar A: Hyperfractionated chemoradio- Morishita E, Nagao M, Sasaki R: In vivo evi-
Radiat Oncol Biol Phys 1998;41:347–353. therapy with carbogen breathing for advanced dence that erythropoietin protects neurons
9 Fein DA, Lee WR, Hanlon AL, Ridge JA, Lan- cancer of the head and neck. Int J Radiat Oncol from ischemic damage. Proc Natl Acad Sci
ger CJ, Curran WJ Jr, Coia LR: Pretreatment Biol Phys 1999;45(3 suppl):377. USA 1998;95:4635–4640.
hemoglobin level influences local control and 19 Lavey RS, Dempsey WH: Erythropoietin in- 26 Brines ML, Ghezzi P, Keenan S, Agnello D, de
survival of T1–T2 squamous cell carcinomas of creases hemoglobin in cancer patients during Lanerolle NC, Cerami C, Itri LM, Cerami A:
the glottic larynx. J Clin Oncol 1995;13:2077– radiation therapy. Int J Radiat Oncol Biol Phys Erythropoietin crosses the blood-brain barrier
2083. 1993;27:1147–1152. to protect against experimental brain injury.
Proc Natl Acad Sci USA 2000;97:10526–
10531.

18 Oncology 2002;63(suppl 2):11–18 Harrison/Shasha/Homel


Oncology 2002;63(suppl 2):19–28
DOI: 10.1159/000067148

Raising Hemoglobin: An Opportunity for


Increasing Survival?
Gillian M. Thomas
Department of Radiation Oncology, Obstetrics & Gynecology, University of Toronto,
Toronto-Sunnybrook Regional Cancer Centre, Toronto, Canada

Key Words creased oxygen carrying capacity may lead to increased


Anemia W Hemoglobin W Hypoxia W Angiogenesis W tumor hypoxia, radiation resistance and increased tumor
Cancer W Radiotherapy W Chemotherapy W Surgery W angiogenesis. The interrelationship of low hemoglobin
Prognostic factor W Epoetin alfa levels, hypoxia, tumor angiogenesis and survival is ex-
plored in this article.
Copyright © 2002 S. Karger AG, Basel

Abstract
Although the association between low hemoglobin lev-
els and poorer outcomes in radiation oncology has long Introduction
been recognized, anemia is often overlooked and un-
treated. However, a growing body of clinical evidence In radiation oncology, it is widely accepted that tumor
now indicates that low hemoglobin levels during radia- hypoxia causes radiation resistance. Anemia is also asso-
tion treatment are associated with decreased response ciated with poorer outcomes to radiation. It has been
and survival following radiotherapy. For example, a inferred that there is a causal relationship between low
large Canadian retrospective study in patients receiving hemoglobin levels, the resulting hypoxia and a poor out-
radical radiotherapy for cervical cancer showed that the come of radiotherapy in patients with cancer.
5-year survival rate was 19% higher in those whose Even though hemoglobin levels are monitored at most
hemoglobin during radiation treatment was =12 g/dl radiation oncology centers, anemia is often overlooked by
compared to those with levels ! 12 g/dl. The data suggest radiation oncologists and is frequently only treated if
that clinical trials need to be performed to determine severe. It has been suggested that oncologists do not rou-
whether increasing hemoglobin levels leads to improved tinely treat mild-to-moderate anemia as it is perceived to
local control and survival. The mechanism by which low be clinically unimportant [1] and that patients are often
hemoglobin levels could cause poorer outcomes is not not transfused unless hemoglobin levels fell below 10 g/dl
well understood and needs further elucidation. It is pos- or even 8 g/dl [1, 2]. For example, a US study in 1987
tulated that lower hemoglobin levels resulting in de- showed approximately two-thirds of academic radiation

© 2002 S. Karger AG, Basel Gillian M. Thomas, BSc, MD, FRCPC


ABC 0030–2414/02/0636–0019$18.50/0 GlaxoSmithKline, 7333 Mississauge Road North
Fax + 41 61 306 12 34 Mississauge, Ont. L5N 8L4 (Canada)
E-Mail karger@karger.ch Accessible online at: Tel. +1 905 814 2256, Fax +1 905 814 2100
www.karger.com www.karger.com/ocl E-Mail Gillian.m.thomas@gsk.com
Table 1. Summary of studies which examined the relationship was no consistency in how it was defined (i.e., cut-off
between anemia and outcome (local control B survival) of radiother- hemoglobin levels ranged from 10 to 12.5 g/dl). Neverthe-
apy B chemotherapy in patients with cancer
less, 40 of the 52 (76.9%) studies showed that low hemo-
Tumor site Number of Effect of anemia on outcome, globin levels were adversely related to local control and/or
studies number of studies survival after radical adverse radiotherapy (table 1).
Adverse None
There are two possible explanations, not mutually
exclusive, for the observed relationship between low he-
Bladder 6 6 0 moglobin levels and impaired outcomes with radiothera-
Bronchus 5 4 1 py. First, low hemoglobin levels may be a tumor-related
Cervix 22 19 3 marker for an aggressive cancer. In this scenario, it is
Glioma 1 0 1
unlikely that raising hemoglobin levels will improve the
Head and neck 17 11 6
Prostate 1 0 1 outcome of radiotherapy. The second, and more tradi-
Total 52 (100%) 40 (76.9%) 12 (23.1%) tional, explanation is that there is a causal relationship
between low hemoglobin levels and poor outcome of ther-
apy. With a causal relationship, raising hemoglobin levels
might therefore improve outcome following radiotherapy
[4].
Until recently, clinical evidence to support a causal
oncology departments transfusedpatients only if their he- relationship between low hemoglobin levels and poor out-
moglobin levels were = 10 g/dl [3]. This reluctance to cor- come was relatively limited. A single prospective, ran-
rect anemia was further increased in Canada in the late domized trial, conducted over 30 years ago, was inter-
1980s when the risk of contracting HIV or hepatitis from preted as demonstrating some benefit after correcting
contaminated blood was first recognized. anemia during radiotherapy in patients with cervical can-
Although views are changing, there is still much uncer- cer [5]. Pelvic recurrence occurred in 11 of 67 patients
tainty among radiation oncologists about the clinical im- (16.4%) who received transfusions and maintained hemo-
portance of radiotherapy-associated anemia and the exact globin levels 112 g/dl compared with 21 of 68 patients
benefits of increasing hemoglobin levels. However, a (30.9%) who were given transfusions only if their hemo-
growing body of clinical data is gathering in the medical globin levels dropped to !10 g/dl. No differences in sur-
literature which examines the relationship between hemo- vival were noted between the two treatment groups [5].
globin levels and response to radiotherapy in patients Although this study is widely quoted in the medical litera-
with cancer. The present article reviews these data and ture as proof that correcting hemoglobin levels improves
also seeks to explore some of the downstream mecha- outcomes following radiotherapy, the study was under-
nisms, namely tumor hypoxia and angiogenesis, that may powered and had an inconclusive univariate analysis
link low hemoglobin levels with clinical outcome in pa- which did not assess the possible effect of other prognostic
tients with cancer. It also poses questions for future study factors on patient outcome.
that may help to clarify treatment options for this patient
group.
Studies in Patients with Cervical Cancer

Effect of Hemoglobin Levels on Treatment Radiotherapy Alone


Outcome To examine the relationship between anemia and
treatment outcome more rigorously, a large retrospective
Historic Data study using data from seven Canadian radiation oncology
More than 50 studies have investigated the effect of centers between 1989 and 1992 was performed [4]. The
low hemoglobin levels at the start of radiotherapy B che- aim of the study was to examine the prevalence of anemia,
motherapy on outcomes in patients with various cancers its time course, and the effect of anemia and blood trans-
[mostly of the cervix (42%) or head and neck (33%)] fusions on the treatment outcome in 605 patients who had
mainly using univariate analysis. A summary of these radical radiotherapy for cervical cancer.
studies is provided in table 1. It should be noted that, At presentation, approximately one-third of patients
although the term ‘anemia’ was used in all studies, there had hemoglobin levels of =12 g/dl (i.e., below the lower

20 Oncology 2002;63(suppl 2):19–28 Thomas


Table 2. Significance of prognostic factors on outcome of radiothera-
py in 605 patients with cervical cancer: results of a multivariate anal- 1.0
ysis (with permission from Grogan et al. [4])
—– Hb =12.0 g/dl (n = 337)
0.8 - - - Hb <12.0 g/dl (n = 172)
Prognostic factor p value
0.6

Survival
Significant
Stage 0.0001 0.4
Average weekly nadir hemoglobin level 0.0001
Intracavitary treatment 0.0004 0.2 p <0.003
Squamous histology 0.0446
0.0
Nonsignificant 0 1 2 3 4 5 6
Age Year
Presenting hemoglobin level
Radiation dose
Center
Fig. 1. Survival in patients with carcinoma of the cervix receiving
Transfusion
radiotherapy stratified according to hemoglobin level (Hb) at presen-
Transfusion year
tation (reproduced with permission from Grogan et al. [4]).
Treatment volume
Treatment time
Chemotherapy

1.0

0.8
— L – H (n = 25)
— H – H (n = 228)
limit of the normal range for women). Despite this, only
0.6
Survival

25% of patients received blood transfusions because clini- — L – L (n = 140)


cians were more likely to transfuse patients with nadir — H – L (n = 82)
0.4
hemoglobin levels of !10 g/dl. This is demonstrated by
p <0.0002
the fact that most patients with hemoglobin levels of 0.2
!9 g/dl (90%) and !10 g/dl (77%) were transfused, where-
as much fewer patients (3–41%) with hemoglobin levels 0.0
0 1 2 3 4 5 6
between 10 and 12 g/dl received transfusions. In all, four Year
of the seven centers had policies for blood transfusion
often not followed: two recommended transfusions for
patients with hemoglobin levels of !10 g/dl, and one each Fig. 2. Survival in patients withcarcinoma of the cervix according to
hemoglobin levels at baseline and during radiotherapy, where L indi-
for patients with hemoglobin levels of !11 and !12 g/dl
cates hemoglobin levels of ! 12 g/dl and H indicates hemoglobin lev-
[4]. els of 612 g/dl. Results are adjusted for disease stage and for the use
The Canadian study showed that hemoglobin levels at of intracavitary irradiation (reproduced with permission from Gro-
baseline correlated with patient survival (fig. 1). This is gan et al. [4]).
consistent with earlier data indicating a correlation be-
tween anemia and poor prognosis in patients receiving
radiotherapy B chemotherapy (table 1). Using a cut-off
value for hemoglobin levels of 12 g/dl, the Canadian study levels (table 2). The multivariate analysis showed that
reported a 12% greater 5-year survival rate in patients hemoglobin levels during radiotherapy, rather than at pre-
with baseline hemoglobin levels of 612 g/dl than in those sentation, were predictive of outcome of radiotherapy in
with baseline levels of 612 g/dl (p ! 0.003; fig. 1). Hemo- terms of overall survival (p = 0.0001; table 2) [4] second in
globin levels at baseline also had a significant effect on importance only to tumor stage. Average weekly hemoglo-
disease-free survival (p = 0.005) and control of local pelvic bin nadir, which was calculated by averaging the weekly
disease (p = 0.002) according to univariate analysis [4]. nadir hemoglobin levels for each patient, was taken as an
Of note, however, are the results of the multivariate estimate of hemoglobin levels during radiotherapy. Other
analysis from this study which considered patient-, tu- significant prognostic factors for outcome were disease
mor- and treatment-related factors, as well as hemoglobin stage, intracavitary treatment, and squamous histology

Hemoglobin Levels and Outcome in Cancer Oncology 2002;63(suppl 2):19–28 21


Patients
1.0

0.8 Hb
NT
12.0 g/dl
T
0.6

Survival
NT
11.0–11.9 g/dl
T
0.4
NT
<11.0 g/dl
T
0.2 p <0.0001
Fig. 3. Survival in patients with carcinoma
of the cervix according to transfusion status
0.0
and average weekly nadir hemoglobin levels 0 1 2 3 4 5 6
(Hb) during radiotherapy, where T = trans- Year
fused and NT = not transfused (reproduced
with permission from Grogan et al. [4]).

(table 2). Initial hemoglobin levels were not significant show that it is the hemoglobin level attained, rather than
suggesting that the effect of low presenting hemoglobin the use of blood transfusions, that influenced outcome in
levels could be overcome by raising the level during treat- these patients. It also confirmed the prognostic signifi-
ment. cance of hemoglobin levels during radiotherapy [4].
To further examine the differential impact of low It was concluded from the Canadian study that the
hemoglobin levels during treatment versus those at pre- hemoglobin level during radiotherapy is an important
sentation, the survival data were reanalyzed according to prognostic factor, second only to disease stage, in patients
hemoglobin levels both at baseline and during radiothera- with cervical cancer. The survival data from this study
py (fig. 2; n = 475). The analysis showed that patients who generate the hypothesis that maintaining hemoglobin lev-
had low hemoglobin levels during radiotherapy, regard- els above 12 g/dl in patients with cervical cancer can
less of their baseline hemoglobin levels, had a significantly improve the response to radiotherapy. The study further
poorer rate of survival than those whose hemoglobin lev- showed that the mechanism by which hemoglobin levels
els were maintained greater than 12 g/dl during radiother- are maintained (i.e., transfusion) is not important, but
apy (fig. 2). The 5-year survival rates for those with low rather the hemoglobin level that is attained during radio-
hemoglobin levels during radiotherapy were 51% or less therapy.
compared with rates of at least 70% in patients who had
high hemoglobin levels during radiotherapy (fig. 2). The Concurrent Radiotherapy and Chemotherapy
relapse rates in patients with low hemoglobin levels dur- Since the study by Grogan et al. [4] was completed,
ing radiotherapy (56 and 60%) were almost double those concurrent cisplatin-based chemotherapy and radiothera-
of patients with high hemoglobin levels during therapy py has emerged as the treatment of choice for patients
(32 and 33%). Interestingly, patients with high hemoglo- with advanced cancer of the cervix [6] as for many other
bin levels during radiotherapy also showed significant epithelial cancers.
reductions in both pelvic (p ! 0.0001) and extrapelvic fail- A recent study by Pearcey et al. [7], however, showed
ure rates (p ! 0.0006) [4]. no benefit for the addition of concurrent cisplatin to
Finally, the study examined whether raising hemoglo- radiation. Contrary to the previous trials in patients with
bin levels with blood transfusions influenced the outcome cervical cancer, which demonstrated a survival benefit for
of radiotherapy (fig. 3). A significant stepwise increase in chemoradiotherapy versus radiotherapy alone [8–12],
overall survival was observed with increasing hemoglobin Pearcey et al. [7] observed similar 3- and 5-year survival
levels during radiotherapy (fig. 3; p ! 0.0001). Survival rates with concurrent cisplatin and radiotherapy versus
rates were not significantly different between patients radiotherapy alone in patients with advanced cervical car-
who attained a given hemoglobin level spontaneously and cinoma. While there are many possible explanations for
those who received blood transfusions (fig. 3). These data the lack of benefit observed with chemoradiotherapy in

22 Oncology 2002;63(suppl 2):19–28 Thomas


Fig. 4. Distribution of patients with ad-
vanced cervical cancer according to decrease
in hemoglobin levels (Hb) during therapy.
Patients were randomized to receive pelvic
radiotherapy alone (n = 126) or radiotherapy
plus cisplatin 40 mg/m2 weekly (n = 127) (re-
produced with permission from Pearcey et
al. [7]).

this trial, one reason may have been a differential drop in therapy in a cancer type other than cervical cancer. Some
hemoglobin levels during therapy (fig. 4) between treat- have postulated that the poor results in anemic patients
ment groups. As would be expected, decreases in hemo- are a result of the association between anemia and large
globin levels were found to be significantly greater in tumor volumes and development of distant metastases.
patients receiving chemotherapy plus radiotherapy versus Since laryngeal cancer does not have these characteristics,
those receiving radiotherapy alone (fig. 4). Thus, is it pos- these data add weight to the suggestion that the relation-
sible that the decreases in hemoglobin levels during thera- ship between anemia and outcome is not solely tumor-
py abrogated the beneficial effects of chemotherapy in related; the hypothesis generated is that some decrement
this study. in response to therapy may occur if the hemoglobin level
is low during treatment.

Studies in Patients with Head and Neck Cancer


Effect of Hypoxia on Treatment Outcome
Several studies have looked at the impact of anemia
on clinical outcome following radiotherapy in patients Hypoxia is a characteristic feature of solid tumors
with head and neck cancer (for review see Kumar 2001) which is thought to occur when tumor growth exceeds the
[13]. As an example, van Acht et al. [14] observed that ability of the local microvasculature to supply oxygen. It is
the 10-year rate of disease-free survival was significantly thought that approximately 60% of locally advanced
lower in patients with laryngeal cancer (n = 306) whose squamous cell cervical carcinomas contain hypoxic and/
hemoglobin levels were below normal (!8.5 mmol/l in or anoxic areas of tissue [15]. Resistance to treatment and
men and !7.5 mmol/l in women) after radiotherapy accelerated tumor growth and progression occur as a
than in those with normal hemoglobin levels. Patients result of hypoxia. Hypoxia causes resistance to both
with glottic carcinoma and hemoglobin levels below nor- radiotherapy and chemotherapy.
mal at the start and/or the end of radiotherapy had sig- It is now well established that intratumoral hypoxia
nificantly reduced 10-year disease-free survival rates has a negative effect on locoregional control in patients
(p = 0.009 and 0.0012, respectively), whereas below-nor- receiving definitive radiotherapy for head and neck or
mal hemoglobin levels at the end of therapy only were cervical cancer. Table 3 provides a summary of studies
predictive of disease-free survival in those with supra- that measured tumor oxygenation levels directly and cor-
glottic cancers (p = 0.05) [14]. related tumor oxygenation status with locoregional con-
These data are of interest because they confirm the trol following radiotherapy [16–21]. Although the defini-
negative association between low hemoglobin levels at the tions assigned to hypoxia differed between studies, four of
end of treatment and survival following definitive radio- six studies showed a significant increase in local failure

Hemoglobin Levels and Outcome in Cancer Oncology 2002;63(suppl 2):19–28 23


Patients
Table 3. Summary of studies investigating
locoregional control following radiotherapy Study Tumor site Local failure rate (patients) p value
according to tumor oxygenation status in nonhypoxic hypoxic
patients with cervical or head and neck tumors tumors
cancer
Fyles et al. [16] Cervix 4/21 6/10 0.03
Höckel et al. [17] Cervix 4/19 10/23 0.13
Kolstad [18] Cervix 4/21 6/10 0.03
Brizel et al. [19] Head and neck 3/10 11/17 0.09
Gatenby et al. [20] Head and neck 1/19 11/12 !0.001
Nordsmark et al. [21] Head and neck 5/18 11/17 0.03

rate in patients who had hypoxic tumors compared with Table 4. Molecular events linking hypoxia and angiogenesis [25]
those whose tumors were oxygenated (table 3).
Hypoxia-regulated genes (e.g., VEGF, EPO, LDHA, Glut-1)
It has long been recognized that hypoxia adversely
HIF-1· mediates transcriptional response by binding to the hypoxia
affects the sensitivity of tumor cells to many chemothera- responsive elements of genes
peutic agents. Although the precise mechanisms are un- HIF-1· induces VEGF and increases expression and half-life of
known, oxygen is a radiosensitizer and impairs the ability mRNA
to repair DNA damage caused by radiation-induced free Hypoxia leads to loss of p53, increases VEGF and decreases TSP-1
H-ras and V-src (oncoproteins) amplify response to hypoxia and
radicals.
lead to increased VEGF and decreased TSP-1
More recently, attention has focused on the ability of
tumor hypoxia to enhance malignant progression. This is EPO = erythropoietin; HIF-1a = hypoxia-inducible factor 1a;
based on the findings of Höckel et al. [17] who showed LDHA = lactate dehydrogenase A; TSP-1 = thrombospondin-1;
that, following tumor resection in 47 patients with cervi- VEGF = vascular endothelial growth factor.
cal cancer, hypoxic tumors had larger extensions, more
frequent parametrial spread and lymph-vascular involve-
ment compared with oxygenated tumors. It is thought
that hypoxia may drive disease progression through clonal
selection and genome changes (for review see Höckel & although the available data are limited. Obermair et al.
Vaupel 2001) [22], which in turn produces a growth [26] used microvessel density count as a measure of angio-
advantage for tumor cells that are resistant to apoptosis. genesis in patients with stage IB cervical cancer who
Hypoxic tumors may overexpress the suppressor gene underwent surgery. They found that the 5-year overall
p53, a phenotype with a high malignant potential [23]. survival rate was significantly better in patients with a
Hypoxia may also induce changes within the tumor microvessel density of 20/field (n = 102) than in those
cells for the expression of oxygen-dependent proteins, with higher microvessel densities (n = 64) (80.7 versus
such as vascular endothelial growth factor (VEGF), which 63.0%; p ! 0.0001).
stimulate angiogenesis and increase the potential for tu- More recently, Birner et al. [27] showed that the
mor growth and metastases [24]. expression of hypoxia-inducible factor 1a (HIF-1a), a
transcriptional factor that promotes angiogenesis and reg-
ulates genes involved in the response to hypoxia, in-
Effect of Angiogenesis on Treatment Outcome fluenced prognosis in 91 surgically-treated patients with
stage I cancer of the cervix. Once again, patients with
Angiogenesis, the growth of new capillary vessels sup- strong expression of HIF-1a had a significantly poorer
porting tumor growth and progression, is stimulated by overall survival (p = 0.03) and disease-free survival (p !
hypoxia. A summary of molecular events linking angio- 0.0001) compared with those with moderate or no expres-
genesis with intratumoral hypoxia is provided in table 4 sion of HIF-1a.
[25]. Like hypoxia, angiogenesis has also been shown to
influence outcome to surgery in patients with cancer,

24 Oncology 2002;63(suppl 2):19–28 Thomas


Fig. 5. Possible management options to overcome the problems of anemia and hypoxia during radiotherapy. HCRS =
Hypoxic cell radiation sensitizers; HT = hyperthermia; HBO = hyperbaric oxygen.

Anemia, Hypoxia and Angiogenesis: Are They type and individual patient physiology, may also be in-
Linked? volved.
The relationship between anemia and angiogenesis
While there is evidence to suggest a direct association remains poorly understood and data are sparse. A Ger-
between hypoxia and angiogenesis, less is known about man group [30] showed that there was a trend towards
how anemia is linked to hypoxia and angiogenesis (fig. 5). higher serum VEGF levels in cancer patients with low
Anemia may exacerbate intratumoral hypoxia by lower- hemoglobin levels undergoing radiotherapy, suggesting
ing the oxygen carrying capacity of the blood, although the that anemia may stimulate angiogenesis via hypoxia. Pa-
link between the two and its relevance in the clinical set- tients had previously untreated, non-metastatic gyneco-
ting remain controversial [1]. Nordsmark et al. [28] in logic cancer (n = 22), head and neck cancer (n = 14), gas-
Denmark recently showed that there was no relationship trointestinal cancer (n = 13), lung cancer (n = 4) and pros-
between hemoglobin levels and pretreatment tumor oxy- tate cancer (n = 1). In 26 patients with hemoglobin levels
gen partial pressure in 263 patients with head and neck of !13 g/dl, mean serum VEGF levels were 805,656 pg/ml
carcinoma. However, both hemoglobin levels and tumor compared with levels of 438,360 pg/ml in 28 patients with
oxygenation status (pO2 fraction !2.5 mm Hg) were inde- hemoglobin levels of 113 g/dl (p = 0.016) [30].
pendent prognostic factors for overall survival. Likewise
Brizel et al. [29] noted only a weak association between
low hemoglobin levels and poor tumor oxygenation status Correcting Hemoglobin Levels
in patients with head and neck cancer receiving primary
radiotherapy; many patients with higher hemoglobin lev- There are several potentially reversible causes of ane-
els (= 13 g/dl) also had hypoxic tumors. These data suggest mia (such as nutritional deficiencies, chronic blood loss,
that low hemoglobin levels and tumor hypoxia may be and subclinical disseminated intravascular coagulopa-
linked in a complex fashion but other factors, tumor thy), which should be sought and, if possible, corrected in

Hemoglobin Levels and Outcome in Cancer Oncology 2002;63(suppl 2):19–28 25


Patients
patients presenting with anemia. A summary of manage- Future Research
ment options for the prevention and treatment of hypoxia
and anemia in patients with cancer in the radiation onco- Examining the impact of raising hemoglobin levels in
logy setting, some of which are in the early stages of devel- cancer patients and finding ways of overcoming tumor
opment, are summarized in figure 5. hypoxia will be important areas of research over the next
Transfusion of red blood cells is one readily available decade. The specific questions that need to be addressed
management option for patients with anemia (fig. 5), include:
although it carries risks of unwanted transfusion reac- E Does raising hemoglobin levels improve patient sur-
tions, infectious disease transmission, and possibly im- vival?
munomodulation. Concerns about the risks of homolo- E If so, by what mechanisms (i.e., does it improve the
gous blood transfusion, uncertainty about the benefits effectiveness of radiotherapy or chemotherapy or does
and inconvenience, mean that anemia is often under- it actually switch off the molecular events that lead to
treated in patients undergoing radiotherapy unless they tumor growth, progression and metastases)?
are clearly symptomatic or have very low hemoglobin lev- E What constitutes the ‘optimal’ hemoglobin level in
els (!10 g/dl) [2]. patients with cancer? This may depend not only on
Recombinant human erythropoietin is an alternative tumor- and patient-related factors but also on the func-
option that avoids the risks associated with transfusions. tional endpoint being studied.
While several trials have already demonstrated that epoe- In an attempt to answer some of these questions, the
tin alfa (Procrit®; Ortho Biotech Products, LP, Bridgewat- Gynecologic Oncology Group (GOG) has initiated an
er, NJ; Eprex®/Erypo®; Janssen-Cilag and Ortho Biotech international randomized trial in patients with stage IIB–
outside the US) effectively corrects anemia and improves IVA cervical cancer and hemoglobin levels of !13 g/dl.
quality of life in patients with cancer receiving chemo- Patients will receive standard combination therapy of
therapy [31–35], it is only more recently that its use has weekly cisplatin and radiotherapy with or without subcu-
been examined in patients receiving radiotherapy or com- taneous epoetin alfa 40,000 units administered once
bined chemoradiotherapy [36–42]. weekly. Blood transfusions are permitted in the control
For example, a prospective, multicenter trial per- group if hemoglobin levels fall to !10 g/dl and in the epoe-
formed in Austria [40] showed that 84% of 143 patients tin alfa group if hemoglobin levels drop precipitously or
with anemia responded to epoetin alfa, which was ini- are too low to raise to 112 g/dl before chemoradiotherapy
tiated approximately 10 days prior to radiotherapy or che- begins. As well as survival, patient quality of life and eco-
moradiotherapy. Hemoglobin levels increased at a me- nomic outcomes will be evaluated in this trial.
dian rate of 0.37 g/dl/week with epoetin alfa. These data Ancillary studies will also be performed in an attempt
are consistent with previous studies, which demonstrated to elucidate the underlying molecular mechanisms. These
the efficacy of epoetin alfa in cancer patients [36–39, 41, include monitoring cell markers of hypoxia (EF5 or 2-(2-
42]. Furthermore, there are now data to support the use of nitro-1-H-imidazol-I-yl)-N-(2,2,3,3,3-pentafluoropropyl)
epoetin alfa administered once weekly [41, 42], rather acetamide) and angiogenesis [VEGF, thrombospondin-1
than the less convenient 3-times weekly regimen that is and platelet/endothelial cell adhesion molecule-1 (PE-
currently approved for use in anemic cancer patients CAM-1 or CD31)]. There will also be an assessment of the
receiving chemotherapy. prognostic value of DNA-cisplatin adducts taken from
In a multicenter study performed in the US, Shasha et buckle smears.
al. [42] showed that overall patient quality of life, and
energy and activity levels were significantly improved
(p ! 0.05 versus baseline) after subcutaneous epoetin alfa Conclusions
40,000 units was administered once weekly for 16 weeks.
The size of effect observed with epoetin alfa (0.5–0.7) was A growing body of literature now shows that there is a
judged to be representative of a medium to large improve- relationship between low hemoglobin levels and low rates
ment in patient quality of life [42]. of disease control and survival in the radiation oncology
setting. Data suggest that correcting anemia with epoetin
alfa improves quality of life, and we postulate, may
improve response and thus impact survival following
radiotherapy with or without concurrent chemotherapy.

26 Oncology 2002;63(suppl 2):19–28 Thomas


Although the long-term implications of correcting anemia quality of life also. This is particularly relevant given the
have yet to be definitively established, collectively, these aggressive combined modality and high-dose treatment
data suggest that it may be important to maintain adequate strategies commonly used in oncology today. Therefore,
hemoglobin levels in patients receiving radiotherapy. the challenge for the future will be to devise a unified
At present, radiation oncologists are focused on wheth- approach to the management of anemia in patients with
er or not it is possible to improve the control of local dis- cancer, with the aim of improving both disease outcome
ease and survival, but it is important to consider patient and patient quality of life.

References

1 Shasha D, Harrison LB: Anemia treatment and 11 Keys HM, Bundy BN, Stehman FB, Muder- 21 Nordsmark M, Overgaard M, Overgaard J:
the radiation oncologist: optimizing patient spach LI, Chafe WE, Suggs CL, Walker JL, Pretreatment oxygenation predicts radiation
outcomes. Oncology (Huntingt) 2001;15:1486– Gersell D: Cisplatin, radiation, and adjuvant response in advanced squamous cell carcinoma
1496. hysterectomy compared with radiation and ad- of the head and neck. Radiother Oncol 1996;
2 Littlewood TJ: The impact of hemoglobin lev- juvant hysterectomy for bulky stage IB cervical 41:31–39.
els on treatment outcomes in patients with can- carcinoma. N Eng J Med 1999;340:1154– 22 Höckel M, Vaupel P: Biological consequences
cer. Semin Oncol 2001;26(suppl 8):49–53. 1161. of tumor hypoxia. Semin Oncol 2001;28(suppl
3 Poskitt TR: Radiation therapy and the role of 12 Peters WA, Liu PY, Barrett RJ, Stock RJ, 8):36–41.
red blood cell transfusion. Cancer Invest 1987; Monk BJ, Souhami L, Grigsby P, Gordon W, 23 Hlatky L, Tsionou C, Hahnfeldt P, Coleman
5:231–236. Alberts DS: Concurrent chemotherapy and pel- CN: Mammary fibroblasts may influence
4 Grogan M, Thomas GM, Melamed I, Wong vic radiation therapy compared with pelvic breast tumor angiogenesis via hypoxia-induced
FL, Pearcey RG, Joseph PK, Portelance L, radiation therapy alone as adjuvant therapy vascular endothelial growth factor up-regula-
Crook J, Jones KD: The importance of hemo- after radical surgery in high-risk early-stage tion and protein expression. Cancer Res 1994;
globin levels during radiotherapy for carcino- cancer of the cervix. J Clin Oncol 2000;18: 54:6083–6086.
ma of the cervix. Cancer 1999;86:1528–1536. 1606–1613. 24 Brown LF, Berse B, Jackman RW, Tognazzi K,
5 Bush RS, Jenkin RD, Allt WE, Beale FA, Bean 13 Kumar P: Impact of anemia in patients with Manseau EJ, Dvorak HF, Senger DR: In-
H, Dembo AJ, Pringle JF: Definitive evidence head and neck cancer. Oncologist 2000;5(suppl creased expression of vascular permeability
for hypoxic cells influencing cure in cancer 2):13–18. factor (vascular endothelial growth factor) and
therapy. Br J Cancer 1978;37(suppl):302–306. 14 van Acht MJ, Hermans J, Boks DE, Leer JW: its receptors in kidney and bladder carcinomas.
6 Thomas GM: Improved treatment for cervical The prognostic value of hemoglobin and a de- Am J Pathol 1993;143:1255–1262.
cancer – concurrent chemotherapy and radio- crease in hemoglobin during radiotherapy in 25 Blancher C, Harris AL: The molecular basis of
therapy. N Eng J Med 1999;340:1198–1200. laryngeal carcinoma. Radiother Oncol 1992; the hypoxia response pathway: tumour hypoxia
7 Pearcey RG, Brundage M, Drouin P, Jeffrey J, 23:229–235. as a therapy target. Cancer Metastasis Rev
Johnston D, Lukka H, MacLean G, Souhami L, 15 Vaupel P, Kelleher DK, Höckel M: Oxygena- 1998;17(2):187–194.
Stuart G, Tu D: Phase III trial comparing radi- tion status of malignant tumors: Pathogenesis 26 Obermair A, Wanner C, Bilgi S, Speiser P,
cal radiotherapy with and without cisplatin of hypoxia and significance for tumor therapy. Kaider A, Reinthaller A, Leodolter S, Gitsch G:
chemotherapy in patients with advanced squa- Semin Oncol 2001;2(suppl 8):29–35. Tumor angiogenesis in stage IB cervical cancer:
mous cell cancer of the cervix. J Clin Oncology 16 Fyles AW, Milosevic M, Wong R, Kavanagh correlation of microvessel density with surviv-
2002;20:966–972. MC, Pintilie M, Sun A, Chapman W, Levin W, al. Am J Obstet Gynecol 1998;178:314–319.
8 Whitney CW, Sause W, Bundy BN, Malfetano Manchul L, Keane TJ, Hill RP: Oxygenation 27 Birner P, Schindl M, Obermair A, Plank C,
JH, Hannigan EV, Fowler WC, Clarke-Pearson predicts radiation response and survival in pa- Breitenecker G, Oberhuber G: Overexpression
DL, Liao SY: Randomized comparison of tients with cervix cancer. Radiother Oncol of hypoxia-inducible factor 1alpha is a marker
fluorouracil plus cisplatin versus hydroxyurea 1998;48:149–156. for an unfavorable prognosis in early-stage in-
as an adjunct to radiation therapy in stage IIB- 17 Höckel M, Schlenger K, Aral B, Mitze M, vasive cervical cancer. Cancer Res 2000;60:
IVA carcinoma of the cervix with negative Schäffer U, Vaupel P: Association between tu- 4693–4696.
para-aortic lymph nodes: a Gynecologic Onco- mor hypoxia and malignant progression in ad- 28 Nordsmark M, Rudat V, Lartigau E, Stadler P,
logy Group and Southwest Oncology Group vanced cancer of the uterine cervix. Cancer Res Becker A, Adam M, Molls M, Dunst J, Terris
study. J Clin Oncol 1999;17:1339–1348. 1996;56:4509–4515. D, Overgaard J: Hypoxia and hemoglobin as
9 Morris M, Eifel PJ, Lu J, Grigsby PW, Leven- 18 Kolstad P: Intercapillary distance, oxygen ten- prognostic markers of survival in head & neck
back C, Stevens RE, Rotman M, Gershenson sion and local recurrence in cervix cancer. carcinoma after primary radiation therapy. An
DM, Mutch DG: Pelvic radiation with concur- Scand J Clin Lab Invest 1968;106(suppl):145– international multi-center study (abstract 125).
rent chemotherapy compared with pelvic and 157. Eur J Cancer 2001;37(suppl 6):37.
para-aortic radiation for high-risk cervical can- 19 Brizel DM, Sibley GS, Prosnitz LR, Scher RL, 29 Brizel DM, Dodge RK, Clough RW, Dewhirst
cer. N Eng J Med 1999;340:1137–1143. Dewhirst MW: Tumor hypoxia adversely af- MW: Oxygenation of head and neck cancer:
10 Rose PG, Bundy BN, Watkins EB, Thigpen JT, fects the prognosis of carcinoma of the head changes during radiotherapy and impact on
Deppe G, Maiman MA, Clarke-Pearson DL, and neck. Int J Radiat Oncol Biol Phys 1997; treatment outcome. Radiother Oncol 1999;53:
Insalaco S: Concurrent cisplatin-based radio- 38(2):285–289. 113–117.
therapy and chemotherapy for locally ad- 20 Gatenby RA, Kessler HB, Rosenblum JS, Coia 30 Dunst J, Pigorsch S, Hansgen G, Hintner I,
vanced cervical cancer. N Eng J Med 1999;340: LR, Moldofsky PJ, Hartz WH, Broder GJ: Ox- Lautenschlager C, Becker A: Low hemoglobin
1144–1153. ygen distribution in squamous cell carcinoma is associated with increased serum levels of vas-
metastases and its relationship to outcome of cular endothelial growth factor (VEGF) in can-
radiation therapy. Int J Radiat Oncol Biol Phys cer patients. Does anemia stimulate angiogene-
1988;14:831–838. sis? Strahlenther Onkol 1999;175:93–96.

Hemoglobin Levels and Outcome in Cancer Oncology 2002;63(suppl 2):19–28 27


Patients
31 Littlewood TJ, Bajetta E, Nortier JWR, Ver- 35 Glaspy J, Bukowski R, Steinberg D, Taylor C, 40 Hawliczek R, Oismüller R: The effect of sys-
cammen E, Rapoport B: Effects of epoetin alfa Tchekmedyian S, Vadhan-Raj S: Impact of tematic rHu-erythropoietin (Epoietin alpha
on hematologic parameters and quality of life therapy with epoetin alfa on clinical outcomes [sic]) treatment before and during radiotherapy
in cancer patients receiving nonplatinum che- in patients with nonmyeloid malignancies dur- (radio-chemotherapy) in unselected anemic
motherapy: results of a randomized, double- ing cancer chemotherapy in community onco- cancer patients: results of an Austrian multi-
blind, placebo-controlled trial. J Clin Oncol logy practice. J Clin Oncol 1997;15:1218– center observation study (abstract 1465). Eur J
2001;19:2865–2874. 1234. Cancer Clin Oncol 1999;35(suppl 4):S361.
32 Dammacco F, Silvestris F, Castoldi GL, Grassi 36 Lavey RS: Clinical trial experience using eryth- 41 Gabrilove JL, Cleeland CS, Livingston RB, Sa-
B, Bernasconi C, Nadali G, Perona G, De Laur- ropoietin during radiation therapy. Strahlen- rokhan B, Winer E, Einhorn LH: Clinical eval-
enzi A, Torelli U, Ascari E, Rossi Ferrini PL, ther Onkol 1998;174(suppl 4):24–30. uation of once-weekly dosing of epoetin alfa in
Caligaris-Cappio F, Pileri A, Resegotti L: The 37 Dusenbery KE, McGuire WA, Holt PJ, et al: chemotherapy patients: improvements in he-
effectiveness and tolerability of epoetin alfa in Erythropoietin increases hemoglobin during moglobin and quality of life are similar to
patients with multiple myeloma refractory to radiation therapy for cervical cancer. Int J Ra- three-times-weekly dosing. J Clin Oncol 2001;
chemotherapy. Int J Clin Lab Res 1998;28: diat Oncol Biol Phys 1994;29:1079–1084. 19:2875–2882.
127–134. 38 Antonadou D, Cardamakis E, Sarris G, et al: 42 Shasha D, George MJ, Harrison LB: Once-
33 Garton JP, Gertz MA, Witzig TE, Greipp PR, Effect of the administration of recombinant weekly dosing of epoetin alfa increases hemo-
Lust JA, Schroeder G, Kyle RA: Epoetin alfa human erythropoietin in patients with pelvic globin and improves quality of life in anemic
for the treatment of the anemia of multiple malignancies during radiotherapy. Radiother cancer patients receiving radiation therapy
myeloma. A prospective, randomized, placebo- Onol 1998;48:S122. either concomitantly or sequentially with che-
controlled, double-blind trial. Arch Intern Med 39 Glaser CM, Millesi W, Kornek GV, Lang S, motherapy (poster). Presented at the American
1995;155:2069–2074. Schull B, Watzinger F, Selzer E, Lavey RS: Society of Hematology (ASH), 3 Dec 2000, San
34 Demetri GD, Kris J, Wade J, Degas L, Celia D: Impact of hemoglobin level and use of recom- Francisco (CA).
Quality-of-life benefit in chemotherapy pa- binant erythropoietin on efficacy of preopera-
tients treated with epoetin alfa is independent tive chemoradiation therapy for squamous cell
of disease response or tumor type: results from carcinoma of the oral cavity and oropharynx.
a prospective community oncology study. J Int J Radiat Oncol Biol Phys 2001;50(3):705–
Clin Oncol 1998;16:3412–3425. 715.

28 Oncology 2002;63(suppl 2):19–28 Thomas


Oncology 2002;63(suppl 2):29–38
DOI: 10.1159/000067145

New Chemotherapeutic Agents:


Update of Major Chemoradiation Trials
in Solid Tumors
Walter J. Curran
Department of Radiation Oncology, Jefferson Medical College, Philadelphia, Pa., USA

Key Words Introduction


Radiotherapy W Chemoradiation W Solid tumors W
Locoregional control W Combined modality therapy Combined modality therapy has been instituted for the
treatment of several types of unresectable solid tumors,
with various chemotherapeutic agents used either sequen-
Abstract tially or concurrently with radiation. Some of these com-
The institution of combined modality therapy for unre- bination therapies have resulted in significant improve-
sected solid tumors has resulted in significant improve- ments in tumor control and survival benefit compared
ments in tumor control and survival benefit compared with radiotherapy (RT) alone. For example, Cancer and
with radiotherapy (RT) alone. A number of chemothera- Leukemia Group B (CALGB) 8433 was the first major
py agents that can enhance the effectiveness of RT, such chemoradiotherapy trial to demonstrate a significant sur-
as cisplatin and 5-fluorouracil, are now considered stan- vival advantage with sequential chemotherapy and radia-
dard treatment for patients with a number of cancer tion in patients with inoperable stage III non-small-cell
types. There is growing interest in a number of addition- lung cancer (NSCLC) [1]. These results were subsequently
al agents that have also been found to have radiosensi- confirmed in the same patient population in a phase III
tizing ability. These include paclitaxel, docetaxel, irinote- study carried out by the Radiation Therapy Oncology
can, gemcitabine, and vinorelbine, as well as biologic Group (RTOG) [2].
agents. Other agents may be of value because they act to A number of chemotherapy agents, including radiosen-
counter dose-limiting toxicities associated with RT. This sitizing agents such as cisplatin and 5-fluorouracil (5-FU),
article provides an update of some important, recently that can be used in conjunction with RT have been identi-
completed and ongoing clinical trials evaluating novel fied and found to be effective in enhancing tumor control
chemoradiation protocols, with examples taken primari- and/or improving survival rates in clinical trials. There
ly from studies conducted by the Radiation Therapy has since been growing interest in a number of agents that
Oncology Group (RTOG). Theoretical approaches to the have also been found to have radiosensitizing ability,
development of new agents and combined modality reg- including paclitaxel, docetaxel, irinotecan, gemcitabine,
imens are also discussed. and vinorelbine, as well as agents with other antitumor
Copyright © 2002 S. Karger AG, Basel activities, including the antiangiogenesis drugs [3–5].
Other agents, such as recombinant human erythropoietin

© 2002 S. Karger AG, Basel Walter J. Curran, Jr., MD


ABC 0030–2414/02/0636–0029$18.50/0 Department of Radiation Oncology, Jefferson Medical College
Fax + 41 61 306 12 34 111 S. 11th St.
E-Mail karger@karger.ch Accessible online at: Philadelphia, PA 19107-5097 (USA)
www.karger.com www.karger.com/ocl Tel. +1 215 955 6700, Fax +1 215 955 0412, E-Mail walter.curran@mail.tju.edu
(rHuEPO, epoetin alfa), are of potential value because need for further improvements in the treatment of locally
they may counter dose-limiting toxicities associated with or regionally advanced unresectable tumors.
RT [6–9], and they may potentially increase tumor oxy- Results from several, recent, prospective phase III
genation, and, therefore, the efficacy of RT. trials provide convincing evidence that further optimiza-
This article provides an update of some important tion of RT delivery in the context of combined modality
recently completed and ongoing trials evaluating novel therapy, beyond that evaluated in the CALGB and RTOG
chemoradiation protocols. It also highlights methodologic trials, can result in better tumor control and/or patient
changes in the way new agents and combined modality survival. These studies are described below.
regimens are being developed.
Lung Cancer
RTOG 9410 is a phase III study of 611 patients with
Optimization of Radiotherapy Delivery unresected NSCLC, which compared a standard sequen-
tial protocol (chemotherapy followed by 60 Gy RT/7
The quality and the mode of delivery of RT are of cen- weeks given once daily, initiated at day 50) with two con-
tral importance to the success of combined modality ther- current chemoradiation protocols (RT initiated on day 1
apy. An important hypothesis that underlies the develop- of chemotherapy) [10]. Concurrent RT was administered
ment of new and more effective RT protocols is that either once daily (60 Gy RT/7 weeks) or as hyperfraction-
improvement of radiotherapy delivery will reduce locore- ated RT (69.9 Gy/6 weeks, twice daily). Preliminary me-
gional tumor failures. Consequently, this should lead to dian survival times at a median follow-up time of 40
decreased mortality, and/or decreased treatment-related months for sequential, concurrent RT once daily, and con-
morbidity. current hyperfractionated RT were 14.6, 17.1, and 15.6
Radiotherapy can be optimized in a number of ways. months, respectively, from each patient’s registration. The
These include technical improvements in patient selec- concurrent RT/cisplatin/vinblastine arm had significantly
tion and staging, which involves the use of ever-improv- better survival than the sequential arm with the same
ing imaging techniques, radiotherapy sequencing and agents, with a p-value of 0.038. The rates of grade 3–4 non-
fractionation, image guidance during RT procedures, bra- hematologic toxicity were higher with concurrent vs. se-
chytherapy, the provision of high radiation doses directly quential chemotherapy, but late toxicity rates were similar
to a tumor through the implantation of small radioactive and no differences in grade 5 toxicity rates were noted.
seeds or sources, radiation intensity modulation, and A randomized intergroup study (RTOG 8815) carried
radiation dose escalation. out by Turrisi et al. [11] was aimed at optimizing RT
Until 2000, standard treatment for stage III unresected delivery in patients (n = 417) with small-cell lung cancer
non-small-cell lung cancer has been sequential chemo- (SCLC). After a follow-up of almost 8 years, patients who
therapy and radiation. An example of this is the treatment were given 45 Gy RT, concurrently with cisplatin and eto-
protocol that was evaluated in the CALGB and RTOG poside, twice daily for 3 weeks, had significantly im-
studies in NSCLC mentioned previously [1, 2]. The com- proved median survival rates versus those who received
bination regimen evaluated in both studies involved ad- 45 Gy RT given concurrently only once daily over 5 weeks
ministration of cisplatin, 100 mg/m2 intravenously on (23 months vs. 19 months for patients receiving once-dai-
days 1 and 29, and vinblastine, 5 mg/m2 on days 1, 8, 15, ly vs. twice-daily RT, respectively, p = 0.04). Survival
22, and 29, followed by RT beginning on day 50 (60 Gy). rates at 5 years were 26 and 16% for patients receiving
This was compared with RT alone. Median survival times concurrent twice-daily or once-daily RT, respectively.
for patients treated with this regimen in the CALGB and However, patients receiving twice-daily RT experienced
RTOG studies were 13.7 and 13.2 months, respectively, grade 3 esophagitis significantly more frequently than
versus 9.6 and 11.4 months for patients treated with RT those receiving once-daily concurrent RT (27 vs. 11%, p !
alone. It was concluded by the authors of the CALGB 0.001).
study that the use of sequential chemoradiotherapy in-
creases the projected proportion of 5-year survivors by a Head and Neck Cancer
factor of 2.8 compared with standard RT [1]. Even with The randomized phase III trial RTOG 9111 compared
this improvement, however, approximately 80–85% of concurrent chemoradiation (RT initiated on day 1 of che-
patients treated with sequential chemoradiotherapy will motherapy) versus sequential chemoradiation (RT ini-
be expected to die within 5 years [1]. Therefore, there is a tiated on day 63 after chemotherapy) vs. RT alone, in 547

30 Oncology 2002;63(suppl 2):29–38 Curran


patients with stage 3–4 potentially resectable cancer of the
larynx [12]. The total RT dose was 70 Gy in 7 weeks
(2 Gy/fraction), administered in the same regimen, for all
of the study arms. The results showed that over a 2-year
follow-up period, concurrent chemoradiation was statisti-
cally significantly better for laryngectomy-free survival
time compared with RT alone (66 vs. 52%, respectively,
p = 0.02). Similarly, concurrent chemoradiotherapy was
statistically significantly better than sequential chemora-
diation and RT alone for time to laryngectomy (p =
0.0094 and 0.00035 vs. sequential chemoradiation and
RT alone, respectively) over the same follow-up period.
In another phase III trial (RTOG 9003), Fu et al. [13]
tested the efficacy of hyperfractionation in patients with
locally advanced head and neck cancer, comparing two
types of accelerated fractionation therapy with standard
fractionated RT. A total of 1,113 patients were random-
ized into one of four treatment groups: (1) RT delivered
with standard fractionation at 2 Gy/fraction/day, 5 days/
week, to 70 Gy/35 fractions/7 weeks; (2) hyperfractiona-
tion at 1.2 Gy/fraction, twice daily, 5 days/week, to 81.6
Gy/68 fractions/7 weeks; (3) accelerated fractionation
with split at 1.6 Gy/fraction, twice daily, 5 days/week, to
67.2 Gy/42 fractions/6 weeks including a 2-week rest after
38.4 Gy; or (4) accelerated fractionation with concomi-
tant boost at 1.8 Gy/fraction/day, 5 days/week and 1.5
Gy/fraction/day to a boost field as a second daily treat-
ment for the last 12 treatment days to 72 Gy/42 fractions/
6 weeks. At a median follow-up time of 23 months for all
assessable patients (n = 1,073), those treated with hyper-
fractionation (treatment 2) or accelerated fractionation
with concomitant boost (treatment 4) had significantly
better locoregional tumor control than those treated with
standard fractionation (treatment 1) (54.4 and 54.5% for Fig. 1. RTOG 9003: Phase III trial comparing hyperfractionated RT
treatments 2 and 4, respectively, vs. 46.0% for treatment or accelerated fractionation RT with concomitant boost vs. standard
RT in patients with advanced head and neck cancer. a Twice-daily
1, p = 0.045 and 0.050, respectively) (fig. 1). A trend
(fractionated) RT resulted in significantly better locoregional control
toward improved disease-free survival was also noted for than did standard RT (p = 0.05). b Accelerated fractionation resulted
the same comparisons (37.6 and 39.3% for treatments 2 in significantly better locoregional control than did standard, twice-
and 4, respectively, vs. 31.7% for treatment 1), although daily fractionation (p = 0.05). Follow-up was from the time patients
these differences failed to achieve significance (p = 0.067 were randomized into the study (reproduced with permission from
Fu et al. [13]).
and 0.054, respectively). All three altered fractionation
groups (treatments 2, 3, and 4) had increased grade 3 or
worse acute adverse effects in skin, mucous membranes,
pharynx/esophagus, and larynx compared with the stan- sequential therapy, and in patients with SCLC, RT twice
dard RT group (treatment 1), but there was no difference daily significantly improved survival rates vs. once-daily
in late toxicity (190 days after start of RT). treatment. Clearly, it is important to keep in mind the val-
The results of these studies clearly demonstrate that ue of modifying RT delivery when attempting to optimize
the quality of RT can be modified to improve tumor con- established chemoradiation combinations, and when test-
trol and survival. In patients with NSCLC or laryngeal ing new systemic agents for use in combined modality reg-
cancer, concurrent RT provided a clear benefit over imens.

Update on Chemoradiation Trials in Solid Oncology 2002;63(suppl 2):29–38 31


Tumors
Integration of New Systemic Therapies with In a prospective, randomized study conducted by
Optimized Locoregional Therapy Hanks et al. [16], a total of 1,554 patients with locally
advanced prostate cancer received androgen suppression
A second important hypothesis that forms the basis for therapy (goserelin and flutamide) 2 months before and
the development of novel combined modality therapies is during RT. Patients were then randomized to either no
the notion that the integration of new systemic therapies further therapy or 24 months of additional goserelin
with optimized locoregional RT will decrease cancer- alone; the groups were well matched for stratification and
related mortality. A number of recent trials were designed other baseline variables considered. The investigators
to compare the efficacy of RT with or without systemic found that disease-free survival (54 vs. 34%), local pro-
therapy in patients with prostate cancer (RTOG 8531, gression (6 vs. 13%), and reduction in distant metastasis
8610, 9202), stage III NSCLC (RTOG 8808), laryngeal (11 vs. 17%) were significantly better in patients receiving
cancer (RTOG 9111), esophageal cancer (RTOG 8501), the longer-duration hormonal therapy compared with
and cervical cancer (RTOG 9001). Overall, the results those receiving no further hormonal therapy after radia-
from these trials, described in the following sections, indi- tion. Patients with a Gleason score of 8–10 (more aggres-
cate a significant clinical benefit from the addition of one sive tumors) also showed a statistically significant advan-
or more systemic agents to RT regimens. tage in overall survival compared with the same subset of
patients who did not receive long-term hormone therapy
Prostate Cancer (p = 0.017). It should be noted that long-term androgen
The benefits of combined modality chemoradiotherapy ablation has hematologic toxicity, which may be detri-
were illustrated in the results of three phase III RTOG mental to locoregional control with RT [17]. Given this
studies that evaluated the combination of RT with hor- possibility, these patients may benefit from additional
monal therapy for treatment of prostate cancer (RTOG systemic agents, such as epoetin alfa, to counter treat-
8531, RTOG 8610, RTOG 9202) [14–16]. A total of 977 ment-related anemias. The potential benefit of epoetin
patients with locally advanced prostate cancer (RTOG alfa for enhancing locoregional tumor control as well as
8531) [14] were followed for a median period of 5.6 years. improving patient quality of life has received support
Results indicated that long-term administration of adju- from recent clinical trials [6–9] and is currently under
vant goserelin (initiated at relapse and continued through- investigation in an RTOG phase III trial (9903).
out the follow-up period) to induce androgen suppression,
in addition to standard external-beam RT (n = 477 analyz- Stage III NSCLC
able patients), significantly improved local tumor control The final results of an important randomized phase III
(p ! 0.0001), freedom from distant metastases, and both trial involving 458 patients with unresectable NSCLC
absolute (p = 0.036) and cause-specific (p = 0.019) survival, were published in 2000 (RTOG 88-08) [2]. This study
as compared with patients who received standard external- compared sequential chemoradiation with either hyper-
beam RT alone (n = 468 assessable patients). fractionated or standard RT. Sause et al. evaluated the
Another randomized, phase III trial (RTOG 8610) [15] efficacy of three regimens: (1) chemoradiation, consisting
investigated the effects of androgen ablation with gosere- of 2 months of cisplatin and vinblastine therapy, followed
lin before and during RT for patients with locally ad- by either 60 Gy of radiation at 2.0 Gy per fraction of
vanced prostate cancer vs. RT alone (n = 471). The results radiation delivered once daily (n = 152), (2) hyperfrac-
at an 8-year follow-up demonstrated that combined mo- tionated RT, consisting of 1.2 Gy per fraction of radiation
dality therapy was associated with a significant improve- delivered twice daily to a total dose of 69.6 Gy (n = 152),
ment in local tumor control (30 vs. 42% local failure rate; and (3) standard RT alone (n = 154). Results indicated
p = 0.016), reduction in disease progression (34 vs. 45% that overall survival was statistically significantly better
distant metastasis; p = 0.04), and improvement in surviv- in patients receiving chemoradiotherapy compared with
al (33 vs. 21% disease-free survival; p = 0.004) compared the RT regimens (median survival of 13.2, 12, and 11.4
with RT alone. A subset analysis of patients with a Glea- months for concurrent chemoradiotherapy, hyperfrac-
son score of 2 to 6 (indicating relatively less aggressive tionated RT, and standard RT, respectively, p = 0.04 for
tumors) showed a highly significant improvement in all chemoradiotherapy vs. the RT regimens); the investiga-
endpoints, including overall survival (p = 0.015) in pa- tors did not find a statistically significant difference in
tients treated with combined modality therapy compared survival between the two RT arms (standard vs. hyper-
with those treated with RT alone. fractionated) in this study.

32 Oncology 2002;63(suppl 2):29–38 Curran


Laryngeal Cancer 0.001 for both parameters). These impressive results of
The objective of RTOG 9111, a randomized, phase III disease-free survival and local control were corroborated
study described previously, was to compare the ability of in three important trials involving women with bulky
sequential chemoradiation (cisplatin 100 mg/m2 plus 5- stage IB cervical cancer [20] or locally advanced cervical
FU 1,000 mg/m2/day ! 120 h for 3 cycles, followed by cancer [21, 22], carried out by the Gynecologic Oncology
RT in responding patients) or concurrent chemoradiation Group. In both of these studies, chemoradiation regimens
(cisplatin 100 mg/m2 plus 5-FU on days 1, 22, 43 plus RT) including cisplatin resulted in significantly improved sur-
with that of RT alone, to promote laryngectomy-free sur- vival and reduced risk of disease recurrence as compared
vival in patients with cancer of the larynx (n = 547) [12]. with RT alone.
Although the study failed to establish a significant differ-
ence in survival between sequential chemoradiation and
standard RT alone, concurrent chemoradiation resulted Unique Approaches to Testing Novel Systemic
in significantly better survival than RT alone (66 vs. 52% Therapies With Radiotherapy
after 2 years, respectively, p = 0.02). Moreover, concur-
rent chemoradiation was also significantly better than A review of trials evaluating combined modality thera-
sequential chemoradiation (p = 0.0094) and RT alone (p = pies illustrates some of the ways in which the approach to
0.00035) with regard to time to laryngectomy. These testing combined modality chemoradiation protocols
results not only demonstrate the benefit of concurrent may differ from traditional drug development strategies.
chemoradiation over standard RT for treatment of laryn- It should always be kept in mind that the latest reports of
geal cancer, as assessed by either survival or time to laryn- new systemic agents undergoing testing, or the most
gectomy, but also highlight the importance of optimizing recent modifications in RT delivery, may prove quite
RT delivery in the context of the specific combined valuable for formulating novel combined modality regi-
modality therapy and the form of cancer involved. mens. It is possible that agents that exhibit only minimal
efficacy when used alone may prove useful when com-
Esophageal Cancer bined with RT. Because of this, many new agents should
Long-term follow-up results (of at least 5 years) from a be tested concurrently with RT after their safety has been
randomized, controlled trial conducted from 1985 to established, rather than postpone concurrent testing until
1990 (RTOG 8501) demonstrated that concurrent che- their efficacy when used alone has been demonstrated. In
moradiotherapy (50 Gy/5 weeks plus cisplatin and 5-FU addition, because of the increasing numbers of potentially
at weeks 1, 5, 8, and 11; n = 134) statistically significantly useful systemic agents, and the resultant exponential
increased the survival of patients with squamous cell can- growth in the numbers of possible therapeutic drug com-
cer or adenocarcinoma of the esophagus compared with binations, the results of preclinical studies will likely play
RT alone (64 Gy/6.4 weeks; n = 62). The overall survival an increasing role in decisions on which new agents
rate with combined therapy was 26% compared with 0% should be tested, and in what types of combinations.
for RT alone [18]. It is important to note that the maximum tolerated
dose (MTD) of a systemic agent, or of a RT protocol, may
Cervical Cancer be defined quite differently when these modalities are
RTOG 9001 was a randomized trial that examined the combined because of the potential interactions between
effects of adding chemotherapy with 5-FU and cisplatin the two forms of therapy. There may be quantitative dif-
to treatment with external beam and intracavitary radia- ferences, such as the dosages at which toxicity is seen, or
tion in women with locally advanced cervical cancer [19]. there may be qualitative differences, such as in the types
Of 403 patients followed for a mean duration of 43 of adverse effects that occur. For example, the MTD may
months, estimated cumulative rates of overall survival relate to organ-specific side effects, such as esophagitis,
were 73% among patients receiving combined modality pneumonitis, or proctitis, rather than to hematologic pa-
therapy, compared with 58% for patients receiving RT rameters, when combined modality therapy is adminis-
alone (p = 0.004). Rates of disease-free survival at 5 years tered. Moreover, interactions between the treatment mo-
were also significantly higher among patients receiving dalities render it likely that optimization of RT delivery
chemoradiation (67 vs. 40%; p ! 0.001), and rates of dis- will result in changes in optimal drug schedules, and vice
tant metastases (14 vs. 33%) and locoregional recurrences versa.
(19 vs. 35%) were significantly lower in these patients (p !

Update on Chemoradiation Trials in Solid Oncology 2002;63(suppl 2):29–38 33


Tumors
Optimization of Combined Modality Regimens tumors, including colon, pancreatic, prostate, lung, and
The results of a phase I trial reported by Fossella et al. head and neck cancers, and is also observed in tumor
[23], aimed at identifying the MTD of gemcitabine in neovasculature [24]. These and other data suggest that
patients with NSCLC undergoing thoracic RT, provide an COX-2-mediated angiogenesis plays a major role in tu-
excellent illustration of how the optimization of RT deliv- mor growth. These findings have stimulated initiation of
ery can play an important role when devising a combined a number of trials evaluating COX-2 inhibitors used in
modality regimen. These investigators found that dose- conjunction with RT.
limiting toxicity of gemcitabine occurred at 125 mg/m2 A phase I/II RTOG trial (C-0128) is underway to deter-
when used with a conventional RT regimen, whereas the mine treatment-related toxicity rates in patients with
MTD was 190 mg/m2 when gemcitabine was combined locally advanced carcinoma of the cervix who are being
with 3-dimensional conformal RT. Thus, optimization of treated with a combination of celecoxib, cisplatin, and 5-
the RT protocol with reduction in the radiation volume FU with concurrent pelvic radiation therapy. In addition,
allowed for the delivery of a substantially higher dose of this study is designed to evaluate whether this regimen
chemotherapy. can improve locoregional control rates, distant control,
RTOG L-0017 is an ongoing phase I trial evaluating and/or survival.
gemcitabine, paclitaxel, and RT vs. gemcitabine, carbo-
platin and RT in patients with NSCLC. The study design Vascular Endothelial Growth Factor (VEGF) Blockade
is an example of a technique that is being used within the VEGF is thought to play an important role in tumor
RTOG to test more efficiently integrated treatment mo- angiogenesis. Blockade of its function is, therefore, con-
dalities. The design includes a first schema that involves sidered a treatment target for a number of tumor types,
administration of escalating doses of gemcitabine concur- and is likely to be tested in conjunction with combined
rently with a constant carboplatin dose, with the combi- modality therapy.
nation at each dose level of gemcitabine given to a group
of 6 patients. A second schema involves an escalation of Sugen (SU) 5416
gemcitabine while also escalating paclitaxel in an alternat- SU 5416 is a small molecule that exhibits potent inhi-
ing stepwise fashion, such that the dose of only one of the bition of the fetal liver kinase (flk) receptor tyrosine
drugs is escalated at a time. In both schemata, chemother- kinase, which is the receptor for VEGF. Expression of flk
apy is accompanied by adjuvant thoracic RT at a total is limited to endothelial cells and it appears to play a criti-
dose of 63 Gy in 34 fractions in 7 weeks to affected areas, cal role in angiogenesis [25]. RTOG S-0120 and S-0121
commencing on the first day of chemotherapy. Using this are two ongoing phase I/II studies aimed at evaluating SU
approach, it should be possible to efficiently establish a 5416 as part of a chemoradiotherapy protocol for treat-
MTD for gemcitabine that is specific for its combination ment of low-to-intermediate grade (S-0120) or high-risk,
with either carboplatin or paclitaxel. high-grade (S-0121) soft tissue sarcoma.

Angiostatin
Novel Systemic Agents Undergoing Testing in In a recent phase I trial at Jefferson Medical College in
Chemoradiotherapy Regimens Philadelphia involving a small number of patients with
advanced cancer of the head and neck, prostate, breast,
Substantial gains have been made over the past several and lung, it was determined that the combination of
years in the induction of remissions with the use of che- angiostatin (an antiangiogenesis drug that has antitumor
moradiation therapy. Despite these advances, patient sur- activity) plus radiotherapy is well tolerated and partial
vival rates are still unacceptably low and new treatment local responses were observed. Studies are ongoing to
strategies are needed. Several promising, novel systemic evaluate longer-term treatment at higher doses [26].
agents are currently undergoing evaluation for use in con-
junction with RT. Farnesyl Transferase Inhibitors
Farnesyl transferase inhibitors target the protein en-
Cyclooxygenase-2 Inhibitors coded by the ras oncogene, blocking its membrane anchor-
Cyclooxygenase (COX) catalyzes the synthesis of pros- age, and thereby inhibiting its cell transforming ability [27].
taglandins from arachidonic acid. One form of the en- R11577 is a potent and selective farnesyl transferase inhib-
zyme, COX-2, is overexpressed in a variety of different itor that has shown antitumor activity in animal models,

34 Oncology 2002;63(suppl 2):29–38 Curran


and was found to have an acceptable tolerability profile in ing, which was derived from archived material and infor-
phase I trials (unpublished data). RTOG 0020 is an ongo- mation, suggests that the development of a strategy for
ing phase II trial in patients with locally advanced pan- EGFR blockade would be useful. Moreover, such a strate-
creatic cancer designed to evaluate the one-year survival gy would be most beneficial if used during radiotherapy,
rates of patients treated with paclitaxel, gemcitabine, and when locoregional failure can be targeted, rather than
RT with or without farnesyl transferase inhibitor after radiotherapy, when attempts to influence distant
R115777. metastases would probably be less useful.
The above represents only a small number of the clini-
cal trials and the novel agents currently being evaluated
for use in combined modality therapies. Given the rela- New Standards of Care
tively recent understanding of the potential value of com-
bining chemotherapeutic and RT regimens for patients Although there is still much work to be done, com-
with unresected solid tumors, this approach can be ex- pleted phase III trials carried out by the RTOG, GOG and
pected to further improve outcomes for patients with other groups have made important contributions to defin-
unresectable solid tumors. ing new standards of care in locally advanced cervical
cancer, stage III NSCLC, locally advanced head and neck
and prostate cancer, localized prostate cancer, central ner-
New Therapeutic Strategies Based on Analyses vous system lymphoma, and operable laryngeal cancer.
of RTOG Clinical and Tissue Databases Some of the RTOG studies are listed in table 1.
A number of RTOG studies, also shown in table 1,
The structure of the RTOG and other groups that are have recently completed accrual, and should be providing
involved in evaluating new combined modality therapies data within the next few years. These trials include much-
encourages the development of such treatments from hy- needed prospective evaluations of radiosurgery. The re-
potheses-based analyses of clinical and tissue databases. sults of two major phase III trials will be presented at the
Over the past few years, there has been a large expansion of 2002 American Society for Therapeutic Radiology and
knowledge concerning the biology of cancer. These ad- Oncology (ASTRO) meeting.
vances include new information on the control of cancer
cell growth and growth factor expression, regulation of Concurrent Chemoradiotherapy Now the Standard
necrotic and apoptotic cell death, regulation of angiogene- The adoption of concurrent rather than sequential che-
sis and cell-cell communication, the influence of environ- motherapy exemplifies how a meaningful improvement
mental factors, such as hypoxia, on tumor growth, and in outcome resulted from relatively subtle changes in ther-
identification of markers that are over- or underexpressed apy, including changes in the technique for delivering che-
on cancer cells. By maintaining centralized databases that moradiotherapy. Using progress that has been made in
make information on basic cancer biology widely available the treatment of stage III NSCLC as an example, the sur-
it will be possible to reassess archived information and uti- vival of good performance status patients with unresected
lize proteomic analysis on banked tissue and tumor speci- tumors progressively improved in a number of studies
mens and apply this knowledge to the formulation of new conducted over approximately 11 years (table 2) [12, 28].
combined modality clinical trials. This degree of improvement cannot be explained by stage
An example of this process involves evaluation of cell migration alone (i.e., changes in staging due to a discrep-
markers, such as epidermal growth factor receptor ancy between clinical and pathologic staging or other fac-
(EGFR), on stored tumor tissue that had been obtained tors). As shown in table 3, the administration of concur-
from patients with advanced head and neck cancer rent rather than sequential chemoradiation, in a random-
treated in study RTOG 9003. This was a phase III study ized RTOG study (9410) of patients with NSCLC, re-
that compared hyperfractionation and standard RT pro- sulted in more than a 20% increase in median survival
tocols [13]. Further analysis of the results of this trial time, a statistically significant improvement [10]. A simi-
revealed that there was a statistically significant increase lar result was seen in a Japanese study reported by Furuse
in locoregional failure in cases where the tumor over- et al. [29]. These results support the positioning of concur-
expressed EGFR vs. those without EGFR, resulting in a rent chemoradiotherapy as the current standard of care.
difference in survival. Interestingly, there was no differ- Although the ability to obtain positive results with consis-
ence in the development of distant metastases. This find- tency across studies is encouraging, it is important to note

Update on Chemoradiation Trials in Solid Oncology 2002;63(suppl 2):29–38 35


Tumors
Table 1. Summary of some recently completed and ongoing major chemoradiation trials

Study Treatment evaluated

Completed phase III RTOG studies


9001 Concurrent chemoradiation for locally advanced cervical cancer
9410 Sequential vs concurrent chemoradiotherapy for stage III NSCLC
9003 Hyperfractionated and accelerated fractionated RT vs standard RT for locally
advanced head and neck cancer
9202, 8610 Long- vs short-term adjuvant hormone therapy for locally advanced prostate cancer
8531 Adjuvant hormone therapy plus RT for localized prostate cancer
9111 Sequential or concurrent chemoradiotherapy vs RT alone for operable laryngeal
cancer
Phase III studies with completed accrual
9305, 9508 Radiosurgery for glioma and brain metastases
9501 Chemotherapy for resected head and neck cancer
9408 Androgen ablation for localized prostate cancer
9413 Large-field RT for advanced prostate cancer
9802 Chemotherapy for low-grade glioma
Unreported/active phase III trials for stage III NSCLC (RTOG and other groups)
RTOG 9801 Amifostine as radioprotectant
ECOG 2597 Role of 3 times daily RT
CALGB 39801 Evaluation of induction chemotherapy
R 9309/Intergroup Role of surgery in N2 disease
ECOG Role of thalidomide
SWOG/Intergroup Role of EGFR blockade

CALGB = Cancer and Leukemia Group B; ECOG = Eastern Cooperative Oncology Group; EGFR = Epidermal
growth factor receptor; N2 = mediastinal lymph node metastasis; NSCLC = non-small-cell lung cancer; RT = radio-
therapy; RTOG = Radiation Therapy Oncology Group; SWOG = Southwest Oncology Group.

Table 2. Evidence for improvement over time in the treatment of Table 3. Concurrent vs. sequential chemoradiotherapy for NSCLC –
unresected NSCLC impact on median survival time

Cooperative Group Trial Median survival 3-year Study Median survival time (months)
time (months) survival
concurrent sequential p value
CALGB 8433 RT [1] 9.6 10%
Curran et al. [10] 17.1 14.6 0.03998
CALGB 8433 sequential CRT [1] 13.7 24%
Furuse et al. [29] 16.5 13.3 0.038
RTOG 9104 concurrent CRT 19.6 40%
RTOG 9410 sequential CRT [2] 14.6 31%
NSCLC = Non-small-cell lung cancer.
RTOG 9410 concurrent CRT [2] 17.0 37%
SWOG 9504 concurrent CRT [28] 27 140%

CALGB = Cancer and Leukemia Group B; RT = radiotherapy;


CRT = chemoradiotherapy; NSCLC = non-small-cell lung cancer; Achieving Further Improvement Using
RTOG = Radiation Therapy Oncology Group; SWOG = Southwest-
Combined Modality Regimens
ern Oncology Group.

Based on the results of the most recent phase III trials


in patients with stage III NSCLC treated with concurrent
that there is much room for further improvement in out- chemoradiation, there appears to be a plateau in median
comes. Moreover, with the concurrent protocols, there is survival time of approximately a year and a half. It is like-
substantially greater acute toxicity; work on the develop- ly that continued optimization of patient selection and the
ment of systemic agents to counter this is ongoing. continued development, selection, and implementation

36 Oncology 2002;63(suppl 2):29–38 Curran


of combined therapies will play important roles in any Conclusions
further improvements in survival times.
One area of research that is contributing to improve- Based on the studies described in this review, it is clear
ments in both patient staging and treatment planning that much has been achieved in the quest for new com-
involves new uses of computed tomography (CT) and bined modality regimens that improve outcomes in pa-
related techniques to provide more detailed information tients with cancer. One major problem, however, is the
on target structures. For example, the use of metabolic long duration of patient accrual, which sometimes results
imaging by means of F-18 fluorodeoxyglucose positron in trials that are inadequately powered. Moreover, there is
emission tomography (FDG-PET) can improve tumor very little information available from controlled trials
definition. This helps the clinician to delineate tumor involving low-performance status patients (based on the
extent and determine the areas that will require high-dose ECOG Performance Status Scale) [32]. Staging of patients
therapy, and to monitor the therapeutic response [30]. has been variable across studies, which can lead to an
Recent work has also indicated that the use of FDG-PET uneven assessment of outcomes. Importantly, there has
staging for NSCLC, for example, is a powerful predictor been too much reliance on the stage IV drug pipeline,
of survival, and it should prove to be a valuable resource rather than expending sufficient effort on developing
in treatment planning [31]. drugs that might be of specific value when used in con-
Continued improvements in radiotherapy delivery, in- junction with RT. This last point relates to differences
cluding dose escalation, intensity modulation, develop- between the strategy for developing combined modality
ment of fractionation techniques, enhanced image guid- regimens testing concurrent regimens early in drug devel-
ance, and exploitation of brachytherapy technology will opment rather than the approach used in standard drug
be crucial to achieving further success; likewise, the ongo- development where single agent efficacy is the first pro-
ing search for novel systemic agents without heavy depen- cess.
dence on drugs that have undergone extensive testing Many factors will contribute to improving outcomes
without RT. Because of the possibility of unique interac- for patients with locally advanced lung cancer, as well as
tions between agents with regard to both efficacy and tox- other solid tumors. Testing novel systemic agents with RT
icity profiles, aggressive concurrent testing, as already is critical, and should pave the way to substantial im-
described, should also play a major role. provements in outcomes. Conducting large explanatory,
The use of chemotherapy ‘doublets’ or ‘triplets’, in proof of principle trials that address a single question,
which novel interactions between chemotherapy agents such as to determine the role of surgery or the optimum
and RT might be exploited, or in which novel biologic timing of sequential therapies, may prove more valuable
agents may be combined with other chemotherapy agents, than complicated studies that mask important principles.
is another promising area of research. For example, work Clearly, improving radiation standards and quality assu-
carried out by Ang and others demonstrated that the com- rance is also critical in this context. All of these ap-
bination of EGFR blockade using monoclonal antibody proaches combined will slowly but surely contribute to-
C225, radiosensitization with docetaxel, and RT resulted wards improving the survival of many patients with local-
in a significantly greater delay in tumor growth than did ly advanced solid tumors.
any one or combination of two therapies (unpublished
data). It is likely that in the next several years, what might
be referred to as new trimodality therapy will be devel-
oped for a number of locally advanced malignancies.

References

1 Dillman RO, Herndon J, Seagren SL, Eaton 2 Sause W, Kolesar P, Taylor S IV, Johnson D, 3 Belani CP: Combined modality therapy for un-
WL Jr, Green MR: Improved survival in stage Livingston R, Komaki R, Emami B, Curran resectable stage III non-small cell lung cancer.
III non-small-cell lung cancer: seven-year fol- WJ Jr, Byhardt R, Dar AR, Turrisi A III: Final New chemotherapy combinations. Chest 2000;
low-up of cancer and leukemia group B results of phase III trial in regionally advanced 117:127S–132S.
(CALGB) 8344 trial. J Natl Cancer Inst 1996; unresectable non-small cell lung cancer. Radia- 4 Gaspar LE: Optimizing chemoradiation thera-
88:1210–1215. tion Therapy Oncology Group, Eastern Coo- py approaches to unresectable stage III non-
perative Oncology Group, and Southwest On- small cell lung cancer. Curr Opin Oncol 2001;
cology Group. Chest 2000;117:358–364. 13:110–115.

Update on Chemoradiation Trials in Solid Oncology 2002;63(suppl 2):29–38 37


Tumors
5 Schiller JH: Novel therapies in lung cancer 14 Lawton CA, Winter K, Murray K, Machtay M, 23 Fossella FV, Zinner RG, Komaki R, Lu R, Lee
management. Oncology 2001;61(suppl 1):1–2. Mesic JB, Hanks GE, Coughlin CT, Pilepich JS, Glisson BS, Pisters KM, Stevens CW, Kurie
6 Glaser CM, Millesi W, Kornek GV, Lang S, MV: Updated results of the phase III Radiation JM, Kies MS, Khuri FR, Kelly J, Liao Z, Hong
Schull B, Watzinger F, Selzer E, Lavey RS: Oncology Group (RTOG) trial 85-31 evaluat- WK: Gemcitabine with concurrent chest radia-
Impact of hemoglobin level and use of recom- ing the potential benefit of androgen suppres- tion followed by consolidation chemotherapy
binant erythropoietin on efficacy of preopera- sion following standard radiation therapy for with gemcitabine plus cisplatin: a phase I trial
tive chemoradiation therapy for squamous cell unfavorable prognosis carcinoma of the pros- for patients stage III non-small cell lung cancer
carcinoma of the oral cavity and oropharynx. tate. Int J Radiat Oncol Biol Phys 2001;49: [abstract]. J Clin Oncol 2001;20:1243.
Int J Radiat Oncol Biol Phys 2001;50:705– 937–946. 24 Masferrer JL, Leahy KM, Koki AT, Zweiferl
715. 15 Pilephich MV, Winter K, John MJ, Mesic JB, BS, Settle SL, Woerner BM, Edwards DA,
7 Harrison LB, Shasha D, Shiaova L, White C, Sause W, Rubin P, Lawton C, Machtay M, Flickinger AG, Moore RJ, Seibert K: Antian-
Ramdeen B, Portenoy R: Prevalence of anemia Grignon D: Phase III radiation therapy oncolo- giogenic and antitumor activities of cyclooxy-
in cancer patients undergoing radiation thera- gy group (RTOG) trial 86-10 of androgen de- genase-2 inhibitors. Can Res 2000;60:1306–
py [abstract]. Proc Am Soc Clin Oncol 2000;19: privation adjuvant to definitive radiotherapy 1311.
1851a. in locally advanced carcinoma of the prostate. 25 Fong TAT, Shawver LK, Sun L, Tang C, App
8 Shasha D, Belani CP, George MJ, Harrison LB: Int J Radiat Oncol Biol Phys 2001;50:1243– H, Powell TJ, Kim YH, Schreck R, Wang X,
Epoetin alfa once weekly increases hemoglobin 1252. Risau W, Ullrich A, Hirth KP, McMahon G:
and improves quality of life in anemic lung 16 Hanks GE, Lu J, Machtay M, Venkatesan V, SU5416 is a potent and selective inhibitor of
cancer patients receiving chemotherapy with or Pinover W, Byhardt R, Rosenthal SA: RTOG the vascular endothelial growth factor receptor
without concomitant or sequential radiation Protocol 92-02: a phase III trial of the use of that inhibits tyrosine kinase catalysis, tumor
therapy [abstract]. J Clin Oncol 2001;20:1243. long-term androgen suppression following vascularization, and growth of multiple tumor
9 George MJ, Vahdat LT, Shasha D, Harrison neoadjuvant hormonal cytoreduction and ra- types. Can Res 1999;59:99–106.
LB: Increased hemoglobin and improved quali- diotherapy in locally advanced carcinoma of 26 Dicker AP, Anne R, Bonanni R, Sidor C, Gu-
ty of life with once weekly epoetin alfa in the prostate [abstract]. Proc Am Soc Clin Oncol bish E, Curran WJ Jr: Phase I trial results of
anemic breast cancer patients receiving con- 2000;19:1284a. recombinant human angiostatin protein and
comitant or sequential chemoradiation [ab- 17 Grogan M, Thomas GM, Melamed I, Wong external beam radiation therapy. American So-
stract]. J Clin Oncol 2001;20:2997. FLW, Pearcey RG, Joseph PK, Portelance L, ciety of Clinical Oncology. 38th Annual Meet-
10 Curran WJ Jr, Scott C, Langer C, Komaki R, Crook J, Jones KD: The importance of hemo- ing, Orlando, Florida, 18–21 May 2002. Ab-
Lee JS, Hauser S, Movsas B, Wasserman TH, globin levels during radiotherapy for carcino- stract 23.
Rosenthal S, Byhardt R, Sause W, Cox J: Phase ma of the cervix. Cancer 1999;86:1528–1536. 27 Gibbs JB, Oliff A: The potential of farnesyl-
III comparison of sequential vs concurrent che- 18 Cooper JS, Guo MD, Herkovic A, Macdonald transferase inhibitors as cancer chemothera-
moradiation for patients with unresected stage JS, Martenson JA, JR, Al-Sarraf M, Byhardt R, peutics. Annu Rev Pharmacol Toxicol 1997;
III non-small cell lung cancer (NSCLC): initial Russell AH, Beitler JJ, Spencer S, Asbell SO, 37:143–166.
report of Radiation Therapy Oncology Group Graham MV, Leichman LL: Chemoradiother- 28 Gandara DR, Lovato LC, Albain KS, Leigh B,
(RTOG) 9410 [abstract]. Proc Am Soc Clin apy of locally advanced esophageal cancer: Lara PN, Crowley JJ, Stelzer K, Livingston RB:
Oncol 2000;19:1891a. long-term follow-up of a prospective random- Prolonged survival in pathologic stage IIIB
11 Turrisi AT, Kim K, Blum R, Sause WT, Liv- ized trial (RTOG 85-01). Radiation Therapy non-small cell lung cancer with concurrent che-
ingston RB, Komaki R, Wagner H, Aisner S, Oncology Group. JAMA 1999;281:1623– moradiotherapy followed by consolidation do-
Johnson D: Twice-daily compared with once- 1627. cetaxel: a phase II study (S9504) of the South-
daily thoracic radiotherapy in limited small- 19 Morris M, Eifel P, Lu J, Levenback C, Grigsby west Oncology Group (SWOG) [abstract]. Proc
cell lung cancer treated concurrently with cis- P, Stevens R, Rotman M, Gershenson D, Am Soc Clin Oncol 2000;19:1916a.
platin and etoposide. New Engl J Med 1999; Mutch D: Pelvic radiation with concurrent che- 29 Furuse K, Hosoe S, Masuda N, Suguira T,
340:265–271. motherapy vs. pelvic and para-aortic radiation Yokota S, Ohbayashi K, Okazaki M, Nakamu-
12 Forastiere AA, Berkey B, Maor M, Weber R, for high-risk cervical cancer: A randomized ra S, Tohda Y, Sawa S, Kamei T, Takada M:
Goepfert H, Morrison WH, Glisson BS, Trotti RTOG clinical trial. New Engl J Med 1999; Impact of tumor control on survival in unre-
A, Ridge JA, Chao C, Peters G, Lee D-J, Leaf 340:1137–1143. sectable stage III non-small cell lung cancer
A, Ensley J: Phase III trial to preserve the 20 Keys HM, Bundy BN, Stehman FB, Muder- treated with concurrent thoracic radiotherapy
larynx: induction chemotherapy and radiother- spach LI, Chafe WE, Suggs CL, Walker JL, and chemotherapy [abstract]. Proc Am Soc
apy vs. concomitant chemoradiotherapy vs. ra- Gersell D: Cisplatin, radiation, and adjuvant Clin Oncol 2000;19:1893a.
diotherapy alone, Intergroup Trial R91–11 [ab- hysterectomy compared with radiation and ad- 30 Choi N, Baumann M, Flentjie M, Kellokumpu-
stract]. J Clin Oncol 2001;20:4. juvant hysterectomy for bulky stage IB cervical Lehtinen P, Senan S, Zamboglou N, Kosmidis
13 Fu KK, Pajak TF, Trotti A, Jones CU, Spencer carcinoma. New Engl J Med 1999;340:1154– P: Predictive factors in radiotherapy for non-
SA, Phillips TL, Garden AS, Ridge JA, Cooper 1161. small cell lung cancer: Present status. Lung
JS, Ang KK: A Radiation Therapy Oncology 21 Rose PG, Bundy BN, Watkins EB, Thigpen JT, Cancer 2001;31:43–56.
Group (RTOG) phase III randomized study to Deppe G, Maiman MA, Clarke-Pearson DL, 31 MacManus MP, Hicks RJ, Ball DL, Kalff V,
compared hyperfractionation and two variants Insalaco S: Concurrent cisplatin-based radio- Matthews JP, Salimen E, Khaw P, Wirth A,
of accelerated fractionation to standard frac- therapy and chemotherapy for locally ad- Rischin D, McKenzie A: F-18 fluorodeoxyglu-
tionation radiotherapy for head and neck squa- vanced cervical cancer. New Engl J Med 1999; cose positron emission tomography staging in
mous cell carcinomas: first report of RTOG 340:1144–1153. radical radiotherapy candidates with nonsmall
9003. Int J Radiat Oncol Biol Phys 2000;48: 22 Whitney CW, Sause W, Bundy BN, Malfetano cell lung carcinoma: powerful correlation with
7–16. JH, Hannigan EV, Fowler WC, Clarke-Pearson survival and high impact on treatment. Cancer
DL, Liao SY: Randomized comparison of 2001;92:886–895.
fluorouracil plus cisplatin versus hydroxyurea 32 American Society of Clinical Oncology. Clini-
as an adjunct to radiation therapy in stage IIB- cal practice guidelines for the treatment of un-
IVA carcinoma of the cervix with negative resectable non-small-cell lung cancer. J Clin
para-aortic lymph nodes: a Gynecologic Onco- Oncol 1997;15:2996–3018.
logy Group and Southwest Oncology Group
study. J Clin Oncol 1999;17:1339–1348.

38 Oncology 2002;63(suppl 2):29–38 Curran

S-ar putea să vă placă și