Sunteți pe pagina 1din 73

2542 PART V  Prevention of Infectious Diseases

245  Active Immunizing Agents

Penelope H. Dennehy • Ian C. Michelow • Michael P. Koster •


Michael A. Smit • Jerome Larkin

Prevention of infectious diseases in children by immunization is one are readily quantifiable and stable in immunobiologic potency. It should
of the outstanding accomplishments of medical science. Children enjoy be easy to administer and not produce disease in the recipient or
better health today because of effective immunization programs, which susceptible contacts. It should induce long-lasting (ideally permanent)
in many countries have markedly diminished the morbidity and mortality immunity that is measurable by available and inexpensive techniques.
of previously common contagious diseases. The striking decline in the It should be free of contaminating and potentially toxic substances,
United States in vaccine-preventable childhood diseases is demonstrated and adverse reactions should be minimal and have minor consequences.
in Table 245.1.4 Immunization programs have led to the global eradication All of these objectives rarely, if ever, are met with the currently available
of smallpox, elimination of measles and poliomyelitis in some regions vaccines because they are neither completely safe nor completely effective.
of the world, and substantial reductions in the morbidity and mortality Partial immunity or undesirable side effects or reactions, or both,
attributed to diphtheria, tetanus, and pertussis. The World Health including rare severe reactions, can occur. Nonetheless, vaccines in
Organization (WHO) estimates that 2 to 3 million child deaths are current use are highly effective and very safe.
prevented by vaccinations annually (http://www.who.int/mediacentre/ All active immunizing agents (i.e., vaccines) contain one or more
factsheets/fs378/en/). antigens that stimulate a protective immunologic response. Some are
To achieve this progress in child health, scientific technology and live attenuated viruses or bacteria; other vaccines consist of killed
medical practice have combined efforts to (1) understand the biology microorganisms or contain inactivated components such as exotoxins
of causal infectious agents, (2) purify these agents and some of their (i.e., toxoids). In some vaccines, the antigen is a highly defined, single
components, (3) develop and test safe and effective vaccines, (4) manu- constituent, such as Haemophilus influenzae type b (Hib) polysaccharide,
facture and administer the vaccines to appropriate segments of the whereas in others, the antigen component is less well defined, such as
population, (5) develop appropriate indications and implement schedules live viruses or whole-cell pertussis vaccines composed of killed Bordetella
for immunizations, and (6) identify necessary contraindications. pertussis organisms.
Infectious diseases can be prevented through immunization by Immunizing agents are administered in suspending fluids such as
stimulating an active immunologic defense (e.g., from humoral antibody) sterile water, saline solution, or complex tissue culture fluid that can
through the administration of antigens, usually before natural exposure contain proteins or other constituents from the medium from which
to an infectious agent (i.e., active immunization) or by temporarily the vaccine was produced (e.g., serum proteins, egg antigens, other
supplying preformed human or animal antibody to persons before or tissue culture–derived antigens). Certain preservatives, stabilizers, or
soon after exposure to certain infectious agents (i.e., passive immuniza- antibiotics are added to some vaccines, and in some cases, the additives
tion). Active immunizations, including the currently available vaccines, can result in hypersensitivity reactions. To enhance immunogenicity,
are discussed in this chapter. The composition of major vaccines and particularly for vaccines containing inactivated microorganisms or
their routes of administration are listed in Table 245.2. their extracted components, adjuvants such as aluminum compounds
are added.
ACTIVE IMMUNOPROPHYLAXIS:
CONSIDERATIONS AND RECOMMENDATIONS Immunization Schedules
The vacinee’s age and timing of immunization are critical for the success
Vaccines of vaccination. The vaccine schedule is based on many factors, including
An ideal immunizing agent should include several characteristics. The the epidemiology of naturally occurring disease, age-specific risk of
agent should be easy to produce in well-standardized preparations that complications caused by the natural disease, anticipated immunologic
CHAPTER 245  Active Immunizing Agents 2543

TABLE 245.1  Reduction in Morbidity of Some Vaccine-Preventable Diseases in the United States
Disease Maximum No. of Cases (y) Cases in 2015 % Decrease
Diphtheria 206,939 (1921) 0 100
Pertussis 265,269 (1934) 20,762 92
Tetanus 1314 (1922–1926) 29 98
Poliomyelitis, paralytic 21,269 (1952) 0 100
Measles 894,134 (1941) 188 >99
Mumps 152,209 (1968) 1329 >99
Rubella 57,686 (1969) 5 >99
Congenital rubella syndrome 20,000 (1964–1965) 1 >99
Haemophilus influenzae type b 20,000 (before 1987) 29 >99
Invasive pneumococcal disease (>5 y) 15,933 (2000) 1177 93
Hepatitis B 21,102 (1990) 3370 84
Varicella 158,364 (1992) 9789 94
Modified from Centers for Disease Control and Prevention. Notice to readers: final 2015 reports of nationally notifiable infectious diseases and conditions. MMWR Morb Mortal Wkly Rep.
2016;651306–21.

TABLE 245.2  Vaccines Available in the United States for Pediatric Use and Routes of Administrationa
Vaccine Type Recommended Route
BCG Live bacteria ID (preferred) or SC
Diphtheria-tetanus (DT, Td) Toxoids IM
DTaP Toxoids and inactivated bacterial components IM
DTaP–hepatitis B–IPV (combination) Toxoids and inactivated bacterial components, recombinant viral IM
antigen, inactivated virus
DtaP-IPV/Hib (combination) Toxoids and inactivated bacterial components, polysaccharide-protein IM
conjugate, inactivated virus
DtaP-IPV (combination) Toxoids and inactivated bacterial components, inactivated virus IM
Hepatitis A Inactivated virus IM
Hepatitis B Recombinant viral antigen IM
Hepatitis A–hepatitis B (combination) Inactivated virus and recombinant viral antigen IM
Hib conjugate Bacterial polysaccharide-protein conjugate IM
Hib–hepatitis B (combination) Bacterial polysaccharide-protein conjugate and recombinant viral antigen IM
Hib conjugate–meningococcal conjugate CY Bacterial polysaccharide-protein conjugate combined with bacterial
(combination) polysaccharide
Human papilloma virus (HPV2, HPV4, and HPV9) Recombinant viral antigens IM
Influenza (IIV) Inactivated viral components IM
Influenza (LAIV) Live attenuated viruses Intranasal spray
Japanese encephalitis Inactivated virus SC
Meningococcal polysaccharide (MPSV4) Bacterial polysaccharide SC
Meningococcal conjugate (MCV4) Bacterial polysaccharide-protein conjugate IM
Meningococcal serogroup B (MenB) Bacterial lipoprotein IM
MMR Live attenuated viruses SC
MMRV Live attenuated viruses SC
Pneumococcal polysaccharide (PPSV23) Bacterial polysaccharide IM or SC
Pneumococcal conjugate (PCV13) Bacterial polysaccharide-protein conjugate IM
Poliovirus (IPV) Inactivated viruses IM or SC
Rabies Inactivated virus IM
Rotavirus (RV1 and RV5) Live attenuated viruses oral
Tdap Toxoids and inactivated bacterial components IM
Tetanus Toxoid IM
Typhoid Bacterial capsular polysaccharide IM
Typhoid Live attenuated bacteria Oral
Varicella Live attenuated virus SC
Yellow fever Live attenuated virus SC
a
Only major childhood vaccines and selected others are included.
BCG, Bacille Calmette-Guérin (tuberculosis); DT, diphtheria-tetanus toxoids (for children <7 y); DTaP, diphtheria-tetanus toxoids–acellular pertussis vaccine (for children <7 y); Hib,
Haemophilus influenzae type b conjugate vaccine; ID, intradermal; IM, intramuscular; IPV, inactivated poliovirus vaccine; MMR, live measles-mumps-rubella viruses vaccine; MMRV, live
measles-mumps-rubella-varicella viruses vaccine; SC, subcutaneous; Td, tetanus-diphtheria toxoid (for children ≥7 y and adults); Tdap, diphtheria–reduced tetanus toxoids–acellular pertussis
vaccine (for children ≥10 y and adults).
2544 PART V  Prevention of Infectious Diseases

response of the host to the antigens, duration of immunity that can be


induced, and recommended ages for routine health care visits. Vaccines TABLE 245.3  Site and Needle Length by Age
are recommended at the youngest age at which significant risk for the for Intramuscular Immunization
natural disease and its complications exist and at which a protective Needle Length, Suggested
immunologic response to the vaccine can occur. Age Group inches (mm) Injection Site
An example is measles vaccine, which in the United States is recom- 5
mended routinely at 12 to 15 months of age because in the first year Neonate (preterm 8 (16) Anterolateral
of life many children have residual, transplacentally acquired maternal and term) and thigh muscle
measles serum antibody that interferes with the antibody response. infants age <1 mo
However, during measles outbreaks or for travel of infants to measles- Term infant, age 1 (25) Anterolateral
endemic areas, measles vaccination is recommended for infants as young 1–12 mo thigh muscle
5
as 6 months because the risk of complications with measles is high Toddlers and children 8 –1 (16–25) Deltoid muscle
among children younger than 1 year.38,471 These infants should be of the arm
vaccinated again at 12 to 15 months of age. Similarly, in countries where 1– 1 14 (25–32) Anterolateral
measles causes significant morbidity and mortality for infants younger thigh muscle
than 9 months, the Global Advisory Group of the WHO’s Expanded Adolescents and
Programme on Immunization (EPI) has recommended giving measles young adults
vaccine to infants as young as 6 months of age.697 Female and male, 1 (25) Deltoid muscle
The recommended doses of vaccine are determined by the number <60 kg of the arm
necessary to achieve a uniform and predictable immunologic response Female, 60–70 kg 1 (25) Deltoid muscle
and to sustain protection. Some immunizing agents require the admin- of the arm
istration of more than one dose for development of an adequate antibody Female, 70–90 kg 1 (25) – 1 12 (38) Deltoid muscle
response and require a booster dose to maintain protection. Examples of the arm
are pertussis, diphtheria, and tetanus vaccines. Intervals between doses Female, >90 kg 1 12 (38) Deltoid muscle
are based on the kinetics of primary and secondary antibody responses. of the arm
Male, 70–118 kg 1 (25) – 1 12 (38) Deltoid muscle
Route of Administration of the arm
An example of the effect of the route of administration on immunologic Male, >118 kg 1 12 (38) Deltoid muscle
response is provided by poliomyelitis vaccines. Inactivated poliovirus of the arm
vaccines given intramuscularly induce systemic immunity through serum
antibody production, but they do not consistently evoke local secretory Modified from American Academy of Pediatrics. Active immunization. In: Kimberlin DW,
Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on
immunoglobulin A (IgA) antibodies in the intestinal tract and thereby Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics;
effectively prevent subsequent transmission of wild-type virus. Because 2015:1–56.
live attenuated oral polio vaccine (OPV) induces optimal intestinal and
systemic antibody, it was the preferred vaccine for routine immunization
of children in the United States against poliomyelitis for 3 decades and
remains the recommended vaccine by the WHO for global eradication.701 immunizing agents cannot replicate in the host, these vaccines must
Vaccines containing adjuvants must be injected deep into the muscle contain an adequate antigenic mass to stimulate the desired immunologic
mass because if they are administered subcutaneously or intradermally, response. Long-lasting immunity with these vaccines requires repeated
they can cause local irritation, inflammation, granuloma formation, or doses. Exceeding the recommended dose can be hazardous because of
necrosis.29,408 excessive local or systemic concentrations of immunizing agents, whereas
Injectable vaccines should be administered in areas unlikely to cause administration of doses smaller than those recommended may result
local neural, vascular, or tissue injury (Table 245.3).29,408 Although the in inadequate response and protection.29,408
upper, outer quadrant of the buttocks has been used as a frequent site
of immunization, this area ordinarily should not be used because the Lapsed Immunizations
gluteal region consists mostly of fat in young children and because of Intervals between multiple doses of an antigen longer than those recom-
potential injury to the sciatic nerve. Ideally, intramuscular injections mended do not affect the antibody responses achieved, provided the
should be given in the anterolateral aspect of the upper part of the thigh immunization series is completed. Restarting the series after interruption
or the deltoid muscle of the upper part of the arm. The anterolateral of the vaccine schedule or giving additional doses is not necessary.
aspect of the thigh is preferred for infants because of its muscle mass
relative to other sites. For older children, the deltoid muscle usually is Simultaneous Administration of Multiple Vaccines
sufficiently large for intramuscular injection. The incidence of significant Because most vaccines can be given simultaneously without impairing
pain in 18-month-old children injected intramuscularly in the thigh is effectiveness or safety, multiple vaccines are given to children concur-
greater than that after deltoid injections and can result in transient rently.392 Simultaneous administration of vaccines is particularly
limping.363 The deltoid is the preferred site for intramuscular administra- important for inadequately immunized children whose return for further
tion of vaccines in children 18 months of age or older, although some immunization is doubtful or for patients with imminent travel plans.
physicians prefer the anterolateral aspect of the thigh for toddlers.29,408 An inactivated vaccine and a live virus vaccine can be administered
Subcutaneous inoculations usually should be given in the thigh of simultaneously at different sites without interference with the immune
infants and the deltoid area of older children. Intradermal vaccines response. Exceptions are yellow fever and cholera vaccines because
should be administered on the volar aspect of the forearm. antibody responses are diminished if they are administered simultane-
Recommended routes for administration of vaccines are provided ously. If possible, administration of these vaccines should be
in their package inserts and are summarized in recommendations for separated by at least an interval of 3 weeks.408 In the case of live virus
immunizations by the Committee on Infectious Diseases of the American vaccines, the immune response to one live virus vaccine can be impaired
Academy of Pediatrics (AAP)29 and the Advisory Committee on if it is given within 4 weeks of another.408 Parenteral live virus vaccines
Immunization Practices (ACIP) of the Centers for Disease Control and not administered on the same day should be given at least 4 weeks
Prevention (CDC).408 apart. This consideration is the basis of the recommended minimal
interval of 1 month (i.e., 4 weeks) between doses of measles
Vaccine Dose vaccine, such as for a previously unimmunized person who is entering
The recommended dose of each immunizing agent is derived from college. Guidelines for spacing live and killed antigen vaccines are given
theoretical considerations and vaccine trials. Because inactivated in Table 245.4.
CHAPTER 245  Active Immunizing Agents 2545

sought through the judicial system by those alleged to have suffered


TABLE 245.4  Guidelines for Spacing the serious vaccine-related sequelae. A marked increase in manufacturers’
Administration of Live and Inactivated Antigens actual and anticipated liability costs and subsequent escalating increases
Antigen Recommended Minimum Interval in the price of vaccines occurred concomitantly. These and other
Combination Between Doses developments, such as threats to the vaccine supply, concerns by parents
about vaccine safety, and recognition of the benefits derived from
Two or more None; can be administered simultaneously or improved coordination and planning of vaccine programs, led to passage
inactivateda at any interval between doses of the 1986 NCVIA and the National Vaccine Injury Compensation
Inactivated plus live None, can be administered simultaneously or Program (https://www.hrsa.gov/vaccinecompensation/index.html), a
at any interval between doses no-fault system to compensate victims of certain presumed vaccine-
Two or more live 28-day minimum interval, if not administered related events.
injectibleb simultaneously The Department of Health and Human Services administers the
a compensation program. Decisions on compensation are made by the
In people with functional or anatomic asplenia, quadrivalent meningococcal conjugate
vaccine (MCV4-D; Menactra) should not be given until at least 4 weeks after all doses of
US Court of Federal Claims and are based on the Vaccine Injury Table,
13-valent pneumococcal conjugate vaccine (PCV13) have been administered because of which was revised since passage of the original legislation in response
interference with the immune response to the PCV13 series because both vaccines are to new findings and analysis by several Institute of Medicine (IOM)
conjugated to diphtheria toxin carrier protein. PCV13 and 23-valent pneumococcal committees. Compensation for injuries occurring after the program’s
polysaccharide vaccine (PPSV23) should not be administered simultaneously and should be effective date of October 1, 1988, is provided by excise taxes on each
spaced at least 8 weeks apart; when both vaccines are indicated, PCV13 should be vaccine. The program has been successful in reducing vaccine-related
administered first, if possible. litigation, stabilizing vaccine prices, and creating a favorable environment
b
An exception is made for some live oral vaccines (e.g., Ty21a typhoid vaccine, oral for the introduction of new vaccines.262
poliovirus vaccine, oral rotavirus vaccine) that can be administered simultaneously or at
any interval before or after inactivated or live parenteral vaccines.
Modified from American Academy of Pediatrics. Active immunization. In: Kimberlin DW,
Vaccine Recommendations and Schedules
Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on In developing recommendations for immunization, many factors are
Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; considered, including the vaccine’s characteristics, scientific knowledge
2015:1–56. about the principles of immunization, assessment of the benefits of
the vaccine, the risk of the disease and its complications, vaccine costs,
and risk of adverse reactions. Changes in relative benefits and risks
necessitate continued review of recommendations.
Record Keeping, Patient Information, Informed In the United States, recommendations for immunization of infants
Consent, and Reporting and children are made by the ACIP of the CDC and the AAP’s Committee
Accurate record keeping by physicians is required, and parents (or on Infectious Diseases. These committees work closely together, and
patients) should keep up-to-date immunization records for their children. in most circumstances, their recommendations are similar. The two
The 1986 National Childhood Vaccine Injury Act (NCVIA) requires committees and the American Academy of Family Practice (AAFP)
that for routinely recommended childhood vaccines, health care providers issue a single vaccine schedule each year. The 2016 schedule for
record in the child’s permanent medical record the date of administration routine administration of childhood vaccines is given in Fig. 245.1 and
of the vaccine, manufacturer, lot number, and name of the health care Table 245.5.
provider administering the vaccines.29 A major change in 1996 was the establishment of a routine preado-
All children and their parents or caregivers should be informed lescent immunization visit at 11 to 12 years of age.125 The dose of adult
about the benefits and risks of the vaccines to be administered. For tetanus-diphtheria–acellular pertussis (Tdap) vaccine should be given
vaccines currently specified in the NCVIA, Vaccine Information State- at that time, as should a dose of the meningococcal conjugate vaccine
ments (VISs) have been prepared by the CDC and must be used by and the first dose of human papillomavirus (HPV) vaccine. At this
vaccine administrators. preadolescent visit, children not previously vaccinated with hepatitis
Informed consent should be obtained before the administration of B, varicella, or the second dose of measles-containing vaccine (or any
vaccines. Some physicians and other health care providers may choose combination of these vaccines) should be given the necessary immuniza-
to obtain the parent’s signature, but current law does not require written tions and scheduled for future visits to receive any vaccines not
consent. An appropriate alternative to written consent is to note in the administered during this visit.
patient’s record that the VISs have been provided and discussed with Other countries have similar national mechanisms for formulating
the parent, patient, or legal guardian.29 immunization schedules and recommendations that are based on the
To increase knowledge about adverse reactions, all temporally associated local epidemiology of diseases and available vaccines. In low-resource
events severe enough to require the patient to seek medical attention should countries, practices are guided by the WHO’s EPI. Current recom-
be reported to the Vaccine Adverse Events Reporting System (VAERS); mendations are listed at the website (http://www.who.int/immunization/
VAERS forms can be obtained by calling 800-822-7967 or by accessing policy/Immunization_routine_table2.pdf?ua=1).
the website (http://vaers.hhs.gov). Health care providers who administer As new vaccines and scientific knowledge become available, vaccine
vaccines are required in the United States to report to the VAERS specific recommendations and schedules are modified and changed. Examples
adverse events in recipients of the vaccines covered by the NCVIA of changes in the past 2 decades include the use of pneumococcal
(Health Resources and Services Administration: http://www.hrsa.gov/ conjugate vaccine beginning at 2 months of age, universal toddler
vaccinecompensation/vaccinetable.html.). This system for reporting adverse hepatitis A immunization, and recommendations for administration
events associated with vaccination was established by the US Department of a second dose of varicella vaccine, and the introduction of acellular
of Health and Human Services to foster recognition of vaccine-related pertussis vaccines for adolescents and adults.
reactions and further study to establish possible causation.
Decreased occurrence of vaccine-preventable infectious diseases has Implementation of Vaccine Programs
resulted in a greater number of adverse events temporally related to In addition to the availability of safe and effective vaccines and appropri-
immunization than cases of disease. Although in some cases, such as ate schedules for their use, effective means of implementation and
vaccine-associated paralytic poliomyelitis, vaccine has been established delivery are necessary for the success of vaccine programs. In the United
as the cause, in other circumstances, such as brain damage alleged to States, high rates of immunization among school-aged children have
be attributed to whole-cell pertussis vaccine, causation by vaccine has been achieved, in part because of public health programs for vaccine
not been proved.14 administration, government support for vaccine purchase, and state
Increased public visibility of vaccine reactions contributed to a laws requiring immunization for school entry. In contrast to rates of
marked increase in vaccine litigation in the 1980s as compensation was approximately 95% or higher among school-aged children, immunization
2546 PART V  Prevention of Infectious Diseases

19–23
Vaccine Birth 1 mo 2 mos 4 mos 6 mos 9 mos 12 mos 15 mos 18 mos 2–3 yrs 4–6 yrs 7–10 yrs 11–12 yrs 13–15 yrs 16–18 yrs
mos

Hepatitis B1 (HepB) 1st dose 2nd dose 3rd dose

Rotavirus2 (RV) RV1 (2-dose st nd See


1 dose 2 dose
series); RV5 (3-dose series) footnote 2

Diphtheria, tetanus, and acellular st nd


1 dose 2 dose 3rd dose 4th dose 5th dose
pertussis3 (DTaP: <7 yrs)

Haemophilus influenzae type b4 See 3rd or 4th dose


1st dose 2nd dose footnote 4
(Hib) See footnote 4

Pneumococcal conjugate5
1st dose 2nd dose 3rd dose 4th dose
(PCV13)

Inactivated poliovirus6
(IPV: <18 yrs) 1st dose 2nd dose 3rd dose 4th dose

Annual vaccination (LAIV or Annual vaccination (LAIV or IIV)


Influenza7 (IIV; LAIV) Annual vaccination (IIV only) 1 or 2 doses IIV) 1 or 2 doses 1 dose only

Measles, mumps, rubella8 (MMR) See footnote 8 1st dose 2nd dose

Varicella9 (VAR) 1st dose 2nd dose

Hepatitis A10 (HepA) 2-dose series, see footnote 10

Meningococcal11 (Hib-MenCY)
6 weeks; MenACWY-D 9 mos; See footnote 11 1st dose Booster

MenACWY-CRM 2 mos)
Tetanus, diphtheria, and acellular
(Tdap)
pertussis12 (Tdap: 7 yrs)

Human papillomavirus13 (2vHPV:


females only; 4vHPV, 9vHPV: (3-dose
males and females) series)

See footnote 11
Meningococcal B11

Pneumococcal polysaccharide5
See footnote 5
(PPSV23)

Range of recommended Range of recommended ages Range of recommended ages Range of recommended ages for non-high-risk No recommendation
ages for all children for catch-up immunization for certain high-risk groups groups that may receive vaccine, subject to
individual clinical decision making
This schedule includes recommendations in effect as of january 1, 2016. Any dose not administered at the recommended age should be administered at a subsequent visit, when indicated and
feasible.The use of a combination vaccine generally is preferred over separate injections of its equivalent component vaccines. Vaccination providers should consult the relevant advisory committee
on immunization practices (ACIP) statement for detailed recommendations, available online at http://www.cdc.gov/vaccines/hcp/acip-recs/index.html. Clinically significant adverse events that follow
vaccination should be reported to the vaccine adverse event reporting system (VAERS) online (http://www.vaers.hhs.gov) or by telephone (800-822-7967). Suspected cases of vaccine-preventable
diseases should be reported to the state or local health department. Additional information, including precautions and contraindications for vaccination, is available from CDC online
(http://www.cdc.gov/vaccines/recs/vac-admin/contraindocations.htm) or by telephone (800-CDC-INFO [800-232-4636]).

This schedule is approved by the advisory committee on immunization practices (http//www.cdc.gov/vaccines/acip), the american academy of pediatrics (http://www.aap.org), the american academy of
family physicians (http://www.aafp.org), and the american college of obstetricians and gynecologists (http://www.acog.org).

NOTE: The above recommendations must be read along with the footnotes of this schedule.

FIG. 245.1  Recommended immunization schedule for people from birth to 18 years of age in the United
States in 2016. An expanded version of the table and its 13 footnotes can be found at https://www.cdc.
gov/vaccines/schedules/hcp/imz/child-adolescent.html. (From Centers for Disease Control and Prevention.
Immunization schedules. http://www.cdc.gov/vaccines/schedules/hcp/child-adolescent.html.)

rates for infants and young children in the 1980s were significantly of pneumococcal conjugate vaccine (PCV).344 Vaccination coverage rates
lower.713 In a survey of 21 primarily urban areas throughout the United in the United States for preschool-aged children consistently have been
States, 11% to 58% (median, 44%) of children who entered school in greater than 70% since 2013.
1991 and 1992 were fully vaccinated by their second birthday. Failure
to immunize young children was a major factor in the outbreaks Vaccine Contraindications, Precautions, and Use
of measles in major urban areas in the United States in 1989 in Special Circumstances
through 1991.634 Recommendations for the use of specific vaccines include contraindica-
The measles epidemic and the recognition of low immunization tions and use in special circumstances, such as immunocompromised
rates prompted a national campaign to achieve the US Public Health patients (from underlying disease or therapy such as high-dose
Service’s goal of a 90% vaccine coverage rate for children by the time corticosteroids) and pregnancy.29,408 Established, generic contraindications
they were 2 years of age. Initiatives have included improved access to are moderate or severe illness, a previous anaphylactic reaction to the
vaccines, education of health care providers in the community, and the specific vaccine, and a severe hypersensitivity reaction (e.g., anaphylaxis)
development of standards for pediatric immunization practice.498 The to a vaccine constituent.
standards have been endorsed by the AAP, AAFP, and other major The decision to defer immunization in a febrile child should be
professional organizations and serve as guidelines to be followed for based on the physician’s assessment of the severity of the illness rather
improving the delivery of vaccines. They include evaluation of the than the degree of fever. Children with minor illness and low-grade
patient’s immunization status at all medical visits, use of valid contra- fever usually should be vaccinated, especially if a child is unlikely to
indications, simultaneous administration of all indicated vaccines, and return promptly for the deferred immunization.
routine audits of the immunization status of patients by providers. Administration of live virus vaccines such as varicella and measles-
These and other initiatives resulted in increasing immunization rates mumps-rubella (MMR) vaccines usually is contraindicated in patients
among young children. with altered immunity. However, the morbidity and mortality rates of
According to the National Immunization Survey, coverage rates for measles and the lack of complications from vaccination of children
children 19 to 35 months of age in 2015 were 72.2% for completion infected with human immunodeficiency virus (HIV) prompted recom-
of the combined vaccine series, which includes four or more doses of mendations that these children, unless significantly immunocompromised,
diphtheria, tetanus toxoids, and acellular pertussis vaccine (DTaP); three receive the MMR vaccine.17,126,471
or more doses of poliovirus vaccine; one or more doses of measles- Because of a theoretical risk to the developing fetus, administration
containing vaccine; full series of Hib vaccine (i.e., three or four doses, of live virus vaccines in most cases is not recommended for pregnant
depending on product type); three or more doses of hepatitis B vaccine women.36,408 However, inadvertent administration of vaccine is not
(HepB); one or more doses of varicella vaccine; and four or more doses necessarily a reason for termination of the pregnancy, and some live
TABLE 245.5  Catch-Up Immunization Schedule for People Age 4 Months Through 18 Years Who Start Late or Are >1 Month Behind—United
States, 2017a

Minimum Age MINIMUM INTERVAL BETWEEN DOSES


Vaccine for Dose 1 Dose 1 to 2 Dose 2 to 3 Dose 3 to 4 Dose 4 to 5
Children 4 Months–6 Years of Age
Hepatitis B1 Birth 4 wk 8 wk and at least 16 wk after first dose — —
Minimum age for the final dose is 24 wk
Rotavirus2 6 wk 4 wk 4 wk2 — —
Diphtheria, tetanus, and acellular pertussis3 6 wk 4 wk 4 wk 6 mo 6 mo3
Haemophilus influenzae type b4 6 wk 4 wk if first dose was administered before 4 wk4 if current age is younger than 12 mo and 8 wk (as final dose); dose —
first birthday first dose was administered before age 7 mo and necessary only for
at least 1 previous dose was PRP-T (ActHib, children 12–59 mo who
Pentacel, Hiberix) or unknown received 3 doses before
8 wk (as final dose) if first dose was 8 wk and age 12–59 mo (as final dose)4 first birthday
administered at age 12–14 mo
No further doses needed if first dose was If current age is younger than 12 mo and first dose
administered at age ≥15 mo was administered at age 7–11 mo or
— If current age is 12–59 mo and first dose was
administered before first birthday and second
dose administered <15 mo or
If both doses were PRP-OMP (PedvaxHIB; Comvax)
and were administered before first birthday
No further doses needed if previous dose was
administered at age ≥15 mo
Pneumococcal5 6 wk 4 wk if first dose administered before first 4 wk if current age is <12 mo and previous dose 8 wk (as final dose); dose —
birthday given at <7 mo of age necessary only for
8 wk (as final dose for healthy children) if 8 wk (as final dose for healthy children) if previous children 12–59 mo of
first dose was administered at or after first dose given between 7–11 mo (wait until at least age who received 3
birthday 12 mo old); or if current age is ≥12 mo and at doses before age 12 mo
least 1 dose was given before age 12 mo or for children at high
No further doses needed for healthy children No further doses needed for healthy children if risk who received 3 —
if first dose administered at age ≥24 mo previous dose administered at age ≥24 mo doses at any age
Continued
CHAPTER 245  Active Immunizing Agents
2547
2548

TABLE 245.5  Catch-Up Immunization Schedule for People Age 4 Months Through 18 Years Who Start Late or Are >1 Month Behind—United
States, 2017a—cont’d

Minimum Age MINIMUM INTERVAL BETWEEN DOSES


Vaccine for Dose 1 Dose 1 to 2 Dose 2 to 3 Dose 3 to 4 Dose 4 to 5
Inactivated poliovirus6 6 wk 4 wk6 4 wk6 6 mo6 (minimum age of —
4 y for final dose).
Measles, mumps, rubella8 12 mo 4 wk — — —
Varicella9 12 mo 3 mo — — —
Hepatitis A10 12 mo 6 mo — — —
PART V  Prevention of Infectious Diseases

Meningococcal11 6 wk 8 wk11 See footnote 11 See footnote 11 —


(Hib-MenCY ≥6 wk; (MenACWY-D ≥9 mo;
MenACWY-CRM ≥2 mo)

Minimum Age MINIMUM INTERVAL BETWEEN DOSES


Vaccine for Dose 1 Dose 1 to Dose 2 Dose 2 to Dose 3 Dose 3 to Dose 4
Children and Adolescents 7–18 Years of Age
Meningococcal11 N/A 8 wk11 — —
(MenACWY-D ≥9 mo; MenACWY-CRM ≥2 mo)
Tetanus, diphtheria; tetanus, diphtheria, and acellular pertussis12 7 y12 4 wk 4 wk if first dose of DTaP/DT was administered before 6 mo if first dose of DTaP/DT
first birthday was administered before
6 mo (as final dose) if first dose of DTaP/DT or Tdap/Td first birthday
was administered at or after first birthday
Human papillomavirus13 9y Routine dosing intervals are recommended13
Hepatitis A10 N/A 6 mo — —
Hepatitis B1 N/A 4 wk 8 wk and at least 16 wk after first dose —
Inactivated poliovirus6 N/A 4 wk 4 wk6 6 mo6
Measles, mumps, rubella8 N/A 4 wk — —
Varicella9 N/A 3 mo if age <13 y — —
4 wk if age ≥13 y
a
The recommendations must be read with the numeric footnotes of this schedule (https://www.cdc.gov/vaccines/schedules/hcp/imz/catchup.html/).
N/A, Not applicable.
From Centers for Disease Control and Prevention. Immunization schedules. http://www.cdc.gov/vaccines/schedules/hcp/child-adolescent.html.
CHAPTER 245  Active Immunizing Agents 2549

virus vaccines, such as those for yellow fever and poliomyelitis, can be
given safely to pregnant women. Inactivated bacterial and viral vaccines BOX 245.1  Misconceptions About Vaccine
such as tetanus toxoids, hepatitis B, and influenza vaccine, which are Contraindications
composed of antigenic components or killed organisms, can and should • Mild acute illness with low-grade fever or mild diarrheal illness in an
be given during pregnancy if indicated. otherwise well child.
In some recipients, vaccines can cause severe reactions, which may
• Current antimicrobial therapy or the convalescent phase of illness.
be a contraindication or precaution to subsequent administration of
the specific vaccine. An example is a child in whom a fever of 40.5°C • Reaction to previous vaccine dose that involved only soreness, redness,
(105°F) or higher develops after receiving DTaP vaccine and for or swelling in the immediate vicinity of the vaccination site or
whom administration of further doses of pertussis-containing vaccine temperature of less than 40.5°C (105°F).
is not indicated in most cases. This recommendation is based on the • Prematurity. The appropriate age for initiating most immunization in the
unproven but reasonable presumption that children who experience prematurely born infant is the usual recommended chronologic age.
adverse reactions after receiving pertussis immunization are at risk for Vaccine doses should not be reduced for preterm infants.
similar reactions of equal or greater magnitude on subsequent • Pregnancy of mother or other household contact. Vaccine viruses in
immunization.188 MMR vaccine are not transmitted by vaccine recipients. Although
Anaphylactic reactions caused by allergenic components of a vaccine varicella vaccine and influenza vaccine viruses have been transmitted
(e.g., gelatin or egg protein in vaccine prepared in embryonated chicken
by a healthy vaccine recipient to contacts, the frequency is rare, only
eggs) have occurred rarely. Vaccines posing a potential risk for egg-
sensitive people include those against measles, mumps, inactivated mild or asymptomatic infection has been reported, and use of vaccine is
influenza, and yellow fever. Before administering vaccines to people not contraindicated by pregnancy of the child’s mother or other
with possible hypersensitivity to vaccine constituents, physicians should household contacts.
review current recommendations for these vaccines. In other circum- • Recent exposure to an infectious disease.
stances, specific immunizations may be contraindicated because of • Breastfeeding. The only vaccine virus that has been isolated from breast
previous reactions and the child’s medical history, such as with DTaP milk is rubella vaccine virus. No evidence indicates that breast milk from
(e.g., evolving neurologic disorders) and MMR (e.g., immune throm- women immunized against rubella is harmful to infants.
bocytopenia associated temporally with vaccination).408 • A history of nonspecific allergies or relatives with allergies.
• Allergies to penicillin or any other antibiotic, except anaphylactic
Misconceptions
reactions to neomycin or streptomycin. These reactions occur rarely, and
Appropriate and safe use of vaccines requires knowledge of the patient’s
relevant medical history, adverse reactions associated with previous none of the vaccines licensed in the United States contains penicillin.
receipt of vaccines, and specific indications and contraindications. • Allergies to duck meat or duck feathers. No vaccine available in the
Without this information, vaccines may be administered inadvertently United States is produced in substrates containing duck antigens.
or not given in circumstances in which immunization is indicated, • Family history of convulsions in persons considered for pertussis or
thereby resulting in missed opportunities for receiving the recommended measles vaccination.
immunization and susceptibility of the child to a preventable disease. • Family history of sudden infant death syndrome in children considered
Examples of common misconceptions concerning contraindication to for DTaP vaccination.
vaccines are given in Box 245.1. • Family history of an adverse event, unrelated to immunosuppression
after vaccination.
International Travel
• Malnutrition.
Foreign travel often is an indication for giving vaccines not routinely
administered to children. The risk of exposure to certain vaccine- DTaP, Diphtheria-tetanus–acellular pertussis; MMR, measles-mumps-rubella.
preventable diseases may be greater than in the United States, and Modified from American Academy of Pediatrics. Active immunization. In: Kimberlin DW,
travelers may be exposed to infections that are uncommon or do not Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on
occur in the United States. Examples include vaccines against Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics;
hepatitis A, typhoid fever, yellow fever, and Japanese encephalitis (JE) 2015:1–56.
(Table 245.6), depending on the location and circumstances of the
person’s visit. Some countries may require yellow fever vaccination for
entry. The second dose of measles vaccine should be given to children
and adolescents who have received only one dose (provided 4 weeks with other medical interventions because in contrast to most pharma-
or more have elapsed since administration of the first dose), irrespective ceutical products that are administered to ill people for curative purposes,
of age because the risk of exposure to measles cases may be substantial vaccines usually are given to healthy people to prevent disease acquisition.
in some foreign countries. Children and adolescents should have received Public tolerance of adverse reactions due to products given to healthy
all vaccines routinely recommended for their age. people, especially healthy infants, is substantially lower than that to
Information on vaccine requirements for international travel is products administered to people who are already ill.
provided in a semiannually revised publication by the CDC, Health The lower tolerance of risk for vaccines necessitates investigating
Information for International Travel678 (https://wwwnc.cdc.gov/travel/ possible causes of rare adverse events after administration of vaccinations
page/yellowbook-home). Information also is available from the CDC more aggressively than would be acceptable for other pharmaceutical
International Travelers Hotline (800-232-4636) or the CDC Traveler’s products. Because vaccination is such a common event, any health
Health website (http://wwwnc.cdc.gov/travel/). problem that occurs after immunization can be attributed to the vaccine.
Health effects reported as being associated with vaccines may be true
Vaccine Safety adverse reactions or may be associated with vaccination only by
Immunizations are among the most cost-effective and widely used public coincidence. Because a temporal relationship alone does not prove
health interventions. Public health recommendations for vaccine causation, cause-and-effect relationships often are impossible to establish.
programs and practices represent a dynamic balancing of risks and Epidemiologic and related studies must be performed to ascertain
benefits. Vaccine safety or risk monitoring is necessary to weigh these the incidence and nature of adverse reactions to vaccines. The studies
factors accurately and adjust vaccination policies accordingly. are important in ensuring a scientific rationale for recommendations
No vaccine is perfectly safe or effective. As the incidence of vaccine- for vaccine use and optimal public and professional acceptance of
preventable diseases is reduced, public concern refocuses from the risk vaccines.
of contracting disease to the health risks associated with vaccines. A The topic of vaccine safety became prominent during the mid-1970s
higher standard of safety typically is expected for vaccines compared with increases in the number of lawsuits filed on behalf of patients
2550 PART V  Prevention of Infectious Diseases

TABLE 245.6  Recommended Immunizations for Travelers to Developing Countries


LENGTH OF TRAVEL
Brief Intermediate Long-Term Residential
Immunizations (<2 wk) (2 wk–3 mo) (>3 mo)
Review and complete age-appropriate childhood schedule + + +
DTaP, poliovirus, pneumococcal, and Haemophilus influenzae type b vaccines may be given
at 4-week intervals if necessary to complete the recommended schedule before
departure. Rotavirus vaccine has maximum ages for the first and last doses;
consideration should be given to the timing of an infant’s travel so that the infant will
be able to receive the vaccine series.
Measles: Infants 6–11 mo of age should receive 1 dose of MMR vaccine before departure.
Two additional doses are given if younger than 12 mo of age at first dose.
Varicella
Hepatitis Aa
Hepatitis Bb
Vaccines against the following diseases should be considered depending on the
geographic area and circumstances of the visit:
Japanese encephalitisc ± ± +
Meningococcal diseased ± ± ±
Rabiese ± + +
Typhoid feverf ± + +
Yellow feverg + + +
a
Indicated for travelers to areas with intermediate or high endemic rates of hepatitis A virus infection.
b
If insufficient time to complete 6-month primary series, an accelerated series can be given.
c
For regions with endemic infection. For high-risk activities in areas experiencing outbreaks, vaccine is recommended, even for brief travel.
d
Recommended for regions of Africa with endemic infection and during local epidemics and required for travel to Saudi Arabia for the Hajj.
e
Indicated for persons at high risk for animal exposure (especially dogs) and for travelers to countries with endemic infection.
f
Indicated for travelers who will consume food and liquids in areas of poor sanitation.
g
For regions with endemic infection.
DTaP, Diphtheria-tetanus–acellular pertussis.
Modified from American Academy of Pediatrics. Immunizations in special clinical circumstances. In: Kimberlin DW, Brady MT, Jackson MA, Long SS, eds.. Red Book: 2015 Report of the
Committee on Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:101–107; Centers for Disease Control and Prevention. Health information for
international travel, 2016: international travel with infants and young children. http://wwwnc.cdc.gov/travel/yellowbook/2016/international-travel-with-infants-children/
vaccine-recommendations-for-infants-children

presumably injured by the diphtheria-tetanus-pertussis (DTP) vaccine.280


Legal decisions were made and damages awarded despite the lack of Reference Sources
scientific evidence to support claims of vaccine injury.280 As a result Several comprehensive sources of information about pediatric vaccines
of the liability, prices soared, and several manufacturers halted production. are available. The AAP publishes The Red Book: Report of the Committee
A shortage of vaccines resulted, and public health officials became on Infectious Diseases every 3 years. The next edition will be published
concerned about the return of epidemic diseases. To reduce liability in 2018. In the interval between editions, the AAP publishes recom-
and respond to public health concerns, Congress passed the NCVIA mendations in its newsletter AAP News and subsequently in Pediatrics.
in 1986. The ACIP issues vaccine recommendations and relevant information
The NCVIA mandates that all health care providers report certain in Morbidity and Mortality Weekly Report. Manufacturers provide product
adverse events that occur after the administration of vaccination with information for each vaccine in the FDA-approved package inserts.
routinely recommended childhood vaccines. As a result, the VAERS
was established by the US Food and Drug Administration (FDA) and VACCINES RECOMMENDED FOR ROUTINE
the CDC in 1990. VAERS provides a mechanism for the collection and ADMINISTRATION
analysis of adverse events associated with vaccines currently licensed
in the United States. Adverse events are defined as health effects occurring Diphtheria Toxoid
after immunization that may or may not be related to the vaccine. The introduction of diphtheria toxoid–containing vaccine in the 1940s
VAERS data are monitored continually to detect previously unknown led to a dramatic reduction in the incidence of diphtheria in the United
adverse events or increases in known adverse events.187 States. From 1980 through 2004, 57 cases of diphtheria were reported
The gaps that exist in the scientific knowledge of rare vaccine adverse in the United States, an average of 2 to 3 per year,405 and only five cases
events prompted the CDC to develop the Vaccine Safety Datalink (VSD) have been reported since 2000. A confirmed case has not been reported
project.185 It involved forming partnerships with four large health in the United States since 2003.31 However, diphtheria remains a
maintenance organizations to monitor vaccine safety continually. VSD potentially significant public health problem. Serologic surveys in the
is an example of a large, linked database and includes information on United States and England have suggested that many adults are not
more than 6 million people. The VSD project enables planned vaccine immune.219,405,449
safety studies and timely investigations of hypotheses. The CDC has Although a study published in 1996 demonstrated that most adults
established an Immunization Safety Office (http://www.cdc.gov/ in the United States do have protective concentrations of serum anti-
vaccinesafety/iso.html) that identifies possible vaccine side effects and toxin,321 data obtained from a national population-based serosurvey
conducts studies to determine whether health problems are caused by published in 2002 revealed that the prevalence of immunity to diphtheria,
vaccines. as determined by the level of diphtheria antitoxin, progressively decreased
CHAPTER 245  Active Immunizing Agents 2551

from 91% among children 6 to 11 years of age to approximately 30% of antitoxin/mL) is reached in more than 95% of vaccinees. Diphtheria
among those 60 to 69 years of age.472 Of the reported cases with known toxoid has been estimated to have a clinical efficacy rate of 97%. Immune
patient age since 1980, 58% were among people 20 years of age or older response to diphtheria toxoid was measured after administration of
and 44% were among people 40 years of age or older. Most cases have each adult Tdap vaccine and compared with that elaborated after Td
occurred in unimmunized or inadequately immunized people. vaccine. Seroprotective antidiphtheria levels, defined as a titer of 0.1 IU/
The current age distribution of cases corroborates the finding of mL or more, and booster response rates to diphtheria were determined
inadequate levels of circulating antitoxin in many adults. Adequate to be noninferior after administration of Tdap compared with Td
immunization has not eliminated the potential for transmission of vaccination.405
Corynebacterium diphtheriae completely because immunization does
not prevent carriage of C. diphtheriae in the nasopharynx or on the Adverse Events
skin.90,405 Other than local reactions of pain and swelling at the site of vaccine
C. diphtheriae requires infection with a virus to acquire the genetic injection, immunization does not cause significant adverse events. These
information for toxin production, which is the primary mediator of local reactions have been attributed to hypersensitivity reactions in
disease. When isolated, strains of C. diphtheriae should be tested for response to the pertussis component and are not a contraindication
toxin production to establish causality in clinical disease. Non–toxin- for administration of further vaccination if otherwise indicated.
producing strains can cause an undifferentiated pharyngitis that is Regarding adverse reactions reported after administration of Tdap,
usually self-limited. Because the organism requires tellurite for growth, vaccination with Boostrix was associated with a statistically higher rate
the microbiology laboratory must be alerted to the fact that diphtheria of moderate to severe headache compared with Td, and vaccination
is suspected when specimens are submitted. Polymerase chain reaction with Adacel was associated with higher rates of mild injection site pain
(PCR) methods can help in making the diagnosis if antibiotics have and low-grade fever compared with the Td vaccine. No serious adverse
been given before obtaining cultures. events have been reported for Boostrix. There have been two reports
C. diphtheriae is further subdivided in to four biotypes: gravis, of serious adverse events, both characterized as neuropathic reactions,
intermedius, mitis, and belfanti. All four biotypes have the potential to in adults, possibly related to having received Adacel (none reported in
produce toxin. Humans are the reservoir for C. diphtheriae and may adolescents), and in both cases, symptoms resolved completely within
be asymptomatic. Transmission occurs by respiratory droplets or several days.90
contaminated fomites. Asymptomatic carriers and those with disease When administered in the third trimester of pregnancy as recom-
can shed the organism for weeks after infection. Shedding is terminated mended by the ACIP, there has been no association with increases in
within 48 hours of initiating appropriate antimicrobial therapy. poor birth outcomes or maternal adverse events.90,243,488,625
As a result of inadequate immunity in adults, infants, and children,
an epidemic of diphtheria occurred in the 1990s throughout the former Indications
Soviet Union, including Russia, Ukraine, and the central Asian republics. Primary immunization consists of five doses of diphtheria toxoid, provided
Case-fatality rates ranged from 3% to 23%.285,334 Diphtheria continues as DTaP or as DT if pertussis is contraindicated.31,90,405 The first three
to be a significant cause of morbidity and mortality in developing doses are administered routinely to children who are 2, 4, and 6 months
countries,285 and it therefore remains a source of possible exposure of age. A fourth dose is administered 6 to 12 months after the third
during travel to endemic countries. Because humans are the only known dose, between 12 to 18 months of age, to maintain adequate antibody
reservoir for C. diphtheriae, universal immunization with a diphtheria concentrations for the ensuing preschool years. For those not immunized
toxoid–containing vaccine is the only effective control measure. in infancy, the initial dose is followed by two doses given 2 and 8 to 14
months later. A single booster dose given when the child is 4 to 6 years
Preparations of age (before school entry) is indicated unless the preceding dose was
Diphtheria toxoid is produced by growing toxigenic C. diphtheriae in given after the fourth birthday. Interruption of the recommended schedule
liquid medium. The filtrate is incubated with formaldehyde to convert or delay in administering subsequent doses during primary immunization
toxin to toxoid and then is adsorbed onto an aluminum salt adjuvant. does not reduce immunity or necessitate restarting the series.
Diphtheria toxoid is available in combination with DTaP for routine In 2005, the FDA licensed two vaccines containing tetanus toxoid,
immunization of infants and children younger than 7 years and for reduced diphtheria toxoid, and acellular pertussis antigens (i.e., Tdap)
adolescents and adults 11 to 64 years of age as a single booster (Tdap). for routine use in adolescents and adults. Tdap is recommended only
Two brands of Tdap are available: Boostrix (approved for people ≥10 for a single dose across all age groups, including as a one-time-only
years of age) and Adacel (approved for people 11 to 64 years of age). substitute for the next scheduled or recommended Td booster in
DTaP and Tdap vaccines do not contain thimerosal as a preservative. individuals who previously completed their primary vaccination series
Diphtheria toxoid also is available as a DTaP–inactivated poliovirus with DTP or DTaP. The preferred age for Tdap vaccination is 11 to 12
(IPV)–hepatitis B combination, a DTaP–inactivated poliovirus (IPV)–Hib years. Adolescents 11 to 18 years of age who completed their primary
conjugate combination, and in combination with tetanus toxoid (i.e., series with DTP or DTaP are encouraged to receive Tdap. Tdap may
DT and Td) for use when pertussis vaccination is contraindicated. No be given without regard to prior immunization history (e.g., those who
diphtheria-only vaccine is available. Hib, pneumococcal, and meningococ- received Td less than 10 years earlier) to a patient for whom pertussis
cal conjugate vaccines containing diphtheria toxoid or CRM197 protein, immunization is indicated.
a nontoxic variant of diphtheria toxin used as an adjuvant, are not Although longer intervals between Td and Tdap could decrease the
substitutes for diphtheria toxoid immunization.31 occurrence of local reactions, the benefits of protection against pertussis
Pediatric formulations (i.e., DT and DTaP) contain an amount of outweigh the potential risk for adverse events.170 Every 10 years thereafter,
tetanus toxoid similar to adult Td, but they contain three to four times a tetanus booster should be provided by Td vaccine. Adolescents between
as much diphtheria toxoid. The concentration of diphtheria toxoid (D) the ages of 11 and 18 years who have never been vaccinated against
for children younger than 7 years per 0.5-mL intramuscular dose of tetanus, diphtheria, or pertussis initially should receive Tdap followed
DTaP or DT vaccine is 6.7 to 25 limit of flocculation (Lf) units, depending by Td for the subsequent two doses at 4 weeks or more and 6 to 12
on the vaccine manufacturer. Vaccines approved for children 7 years months after that.
or older and adults (i.e., Tdap and Td) contain only a fraction of the No pertussis-containing vaccine has been FDA approved for children
diphtheria toxoid (d = 2–2.5 Lf) because of adverse reactions related between the ages of 7 and 10 years. If pertussis vaccine is not contra-
to dose and age.31,90,405 indicated, children 7 through 10 years of age who are not fully vaccinated
(i.e., have not received five doses of DTaP or four doses of DTaP if the
Immunogenicity fourth dose was administered on or after the fourth birthday) should
After a primary series of three properly spaced diphtheria toxoid doses receive a single dose of Tdap to provide protection against pertussis.
in adults or four doses in infants, a protective level of antitoxin (≥0.1 IU If additional doses of tetanus and diphtheria toxoid–containing vaccines
2552 PART V  Prevention of Infectious Diseases

are needed for children 7 through 10 years of age after Tdap vaccine ineffective, particularly in children younger than 18 months who lack
is substituted as the first dose, Td vaccine should be provided for the immunologic memory. Covalent linkage of the purified capsular
subsequent two doses at 4 weeks or more and 6 to 12 months after that polysaccharide, polyribosylribitol phosphate (PRP), to a protein carrier
if indicated for the catch-up series.31,171 Tdap is recommended in this creates a conjugate glycoprotein that is T-lymphocyte dependent and
age group because of its reduced antigen content compared with DTaP, elicits protective antibody in infants and young children and significantly
resulting in reduced reactogenicity.171 Pregnant woman should receive greater concentrations of circulating anti-PRP at all ages than does the
Tdap early in the third trimester of each pregnancy.171 unconjugated polysaccharide. T-cell–dependent antigens involve helper
After exposure to a case of strongly suspected or proven diphtheria, T-lymphocyte activation of a B-cell humoral antibody response.
asymptomatic, previously immunized people should receive a booster T-cell–dependent antigens also are able to prime a booster response
of an age-appropriate diphtheria toxoid–containing vaccine if they have and induce immunologic memory (i.e., anamnestic response).
not received a booster dose of a diphtheria toxoid–containing vaccine The introduction of Hib conjugate vaccines in the United States,
within 5 years. For people 10 years or older, Tdap is preferred over Td first in children at least 18 months of age in 1987 and for routine infant
if a pertussis booster vaccine was not previously received.31 If not previ- immunization in 1990, dramatically decreased the incidence of meningitis
ously immunized, carriers should receive age-appropriate diphtheria and bacteremia caused by Hib. As of 2000, the incidence of invasive
toxoid–containing vaccine (i.e., DTaP [or DT], Tdap, or Td) as soon Hib disease in the United States had decreased by 99% since the prevac-
as identified and should complete the entire series. Patients recovering cine era, with an incidence rate of less than one case per 100,000.143
from diphtheria infection should be immunized because infection does During 2000 through 2012, the average annual incidence of invasive
not confer immunity. disease in young children was lower than the Healthy People 2020 goal
of 0.27 case per 100,000 children.89 The remarkably rapid reduction in
Precautions and Contraindications incidence of disease was partly the result of the ability of the vaccine
The only contraindication to diphtheria toxoid is a history of severe to reduce asymptomatic oropharyngeal colonization in vaccinated
hypersensitivity developing after receipt of a previous vaccine dose. children that also had the indirect effect of reducing exposure and
Vaccination with diphtheria or tetanus toxoid is not associated with an infection in those not immunized (i.e., community or herd immu-
increased risk of convulsions. Local reactions alone do not preclude nity).63,338 Colonization rates have declined from up to 7% in the prevac-
continued use. cine era to less than 1% in the vaccine era.

Haemophilus influenzae Type b Vaccine Preparations


Before the introduction of routine infant and childhood vaccination Three single-antigen Hib conjugate vaccine products and two combina-
against Hib in 1985, this pathogen was the major cause of invasive tion vaccine products that contain Hib conjugate are available
bacterial infections in young children in the United States. It was the in the United States. All Hib vaccines contain polyribosylribitol
most common cause of bacterial meningitis and epiglottitis and a phosphate (PRP), the organism’s polysaccharide capsular antigen.
significant cause of septic arthritis, osteomyelitis, septicemia, occult Three licensed single-antigen Hib conjugate vaccines—PRP-OMP
febrile bacteremia, pneumonia, pericarditis, and cellulitis in children (PedvaxHIB, Merck), PRP-T (ActHIB, Sanofi Pasteur), and PRP-T
younger than 5 years, in whom it caused an estimated 12,000 cases of (Hiberix, GlaxoSmithKline)—are approved for use in early infancy
meningitis and 8000 additional cases of invasive Hib disease annually.198 (Table 245.7).
The cumulative risk for Hib disease was approximately one among Each conjugate vaccine is chemically and immunologically unique.
every 200 US children in the first 5 years of life, with the peak incidence PRP preparations are composed of the type b PRP antigen conjugated
of Hib meningitis occurring among infants between 6 and 12 months to a protein, but they have different protein carriers, sizes of the saccharide
of age. In high-risk populations, such as American Indians and Alaska component, and chemical linkages. The carrier proteins for PRP-OMP
Natives, rates of disease in the absence of immunizations were higher, and PRP-T are an outer membrane protein from Neisseria meningitidis
and a greater proportion of cases of meningitis occurred early in the and tetanus toxoid, respectively. Since July 2000, the entire Hib vaccine
first year of life than in low-risk populations.198,343,680 Although rates of supply for the United States has been thimerosal free. The dose of each
Hib disease among these vulnerable populations have decreased in the Hib conjugate vaccine is 0.5 mL and given intramuscularly.
postvaccine era, they remain 8 to 10 times higher compared with white Two licensed combination vaccines containing Hib are available in
and black children younger than 5 years.89 the United States: DTaP/IPV/PRP-T (Pentacel, Sanofi Pasteur), which
In 2000, before the widespread introduction of Hib vaccines in is approved for immunization at 2, 4, 6, and 15 to 18 months of age,
resource-poor countries, the WHO estimated that at least 8 million and MenCY/PRP-T (MenHibRix, GlaxoSmithKline), a combination
cases of serious disease in children and approximately 371,000 deaths conjugate vaccine approved in 2012 for children 6 weeks to 18 months
were attributable to Hib annually.699 As of 2014, 192 of the 193 WHO of age to prevent meningococcal serogroups C and Y and Hib. Use of
member states (99.5%) had adopted conjugated Hib vaccines in their DTaP/PRP-T (TriHIBit, Sanofi Pasteur) was discontinued in 2011, and
routine immunization programs. As a consequence, invasive Hib disease PRP-OMP/Hep B (Comvax, Merck) was discontinued in 2014.
has been virtually eliminated in many industrialized countries, and its
incidence has declined by more than 90% in resource-poor countries
with effective national immunization programs. However, the WHO
estimates that only 56% of the eligible population globally receives TABLE 245.7  Haemophilus influenzae Type b
three doses of Hib vaccine, and coverage rates are as low as 21% in the
Vaccine Schedules
western Pacific.
Hib remains the most common cause of nonepidemic bacterial VACCINE AGE AT VACCINATION
meningitis among unvaccinated children younger than 1 year. The Type Trade Name 2 mo 4 mo 6 mo 12–15 mo
mortality rate for Hib meningitis treated with appropriate antibiotics
is approximately 4%, and 15% to 30% of survivors have hearing impair- PRP-T ActHIB X (1st) X (2nd) X (3rd) X
ment or severe permanent neurologic sequelae.89 Pentacela X (1st) X (2nd) X (3rd) X
Because most cases of Hib disease occur in infancy, vaccines that Hiberix X (1st) X (2nd) X (3rd) X
induce protection before the age of 6 months are necessary for effective MenHibrixb X (1st) X (2nd) X (3rd) X
control of Hib disease. The Hib polysaccharide capsule is the major PRP-OMP PedvaxHIB X (1st) X (2nd) — X
virulence factor, and bactericidal antibodies directed against the polysac- a
Combination vaccine with DTaP/IPV + PRP-T.
charide antigens are protective. Capsular type b is responsible for more b
Combination vaccine with MenCY + PRP-T.
than 90% of systemic H. influenzae infections. However, purified PRP-OMP, Polyribosylribotol phosphate conjugated to the outer membrane protein
polysaccharide vaccines are poor immunogens and children’s T-lym- complex from Neisseria meningitides; PRP-T, polyribosylribotol phosphate conjugated to
phocyte–independent immunoglobulin M (IgM) response is largely tetanus toxoid.
CHAPTER 245  Active Immunizing Agents 2553

Immunogenicity and Efficacy should be immunized with at least a single dose of any licensed conjugate
Placebo-controlled field trials of Hib conjugate vaccines in infants in Hib vaccine. For previously unimmunized children 5 years or older,
the United States demonstrated almost 100% protection and provided immunization is indicated only if they have an underlying condition
the basis for the initial approval of these vaccines for use in this country. predisposing to Hib disease, such as anatomic or functional asplenia,
In a Navajo population of infants at high risk for Hib disease who sickle cell disease, immunoglobulin deficiency and immunoglobulin
were vaccinated at 2 and 4 months of age with PRP-OMP or placebo, G2 subclass deficiency, early component complement deficiencies, and
vaccine efficacy was 100% at 1 year of age and 93% in total.577 Licensure HIV infection or if they are recipients of hematopoietic stem cell
of PRP-T was based on immunogenicity in a three-dose schedule that transplants or are immunosuppressed because of chemotherapy or
was comparable to that of the other two products. A clinical trial in radiation therapy. More than one dose may be recommended for this
Great Britain found the efficacy of PRP-T comparable to that of population.32,89
PRP-OMP.84,649
Although the data are limited, some evidence suggests that administra- Precautions and Contraindications
tion of the first dose of the Hib vaccine before the child reaches the Adverse reactions to Hib-containing monovalent vaccines are uncommon,
age of 6 weeks may result in immunologic tolerance to the Hib antigen usually mild, and usually resolve within 12 to 24 hours.232,230,282,352 Rates
and reduce the immune response to subsequent doses.473 Therefore, of adverse reactions to Hib combination vaccines are similar to those
Hib vaccines, including combination vaccines that contain Hib conjugate, observed with separately administered vaccines.79,319,493 More complete
should not be given to an infant younger than 6 weeks. information about adverse reactions to a specific vaccine is available
Long-term protection induced by invasive Hib disease in unvaccinated in the package insert for each vaccine.
children is associated with an anti-PRP antibody concentration of greater Vaccination with a Hib-containing vaccine is contraindicated in
than 0.15 µg/mL, whereas concentrations greater than 1.0 µg/mL are infants younger than 6 weeks. Vaccination with a Hib-containing vaccine
considered to be markers of long-term protective immunity in vaccinated is contraindicated for people known to have a severe allergic reaction
children.89 Children who have invasive Hib infection before 24 months to any component of the vaccine. The tip caps of the Hiberix prefilled
of age may remain at risk for another Hib infection, and they should syringes can contain natural rubber latex, and the vial stoppers for
be immunized according to the age-appropriate schedule for unim- Comvax, ActHib, and PedvaxHIB contain natural rubber latex, which
munized children.32 Children who develop invasive disease despite two can cause allergic reactions in people who are latex sensitive. Vaccination
to three Hib vaccines and those with recurrent invasive Hib warrant with these vaccines is therefore contraindicated for those known to
an immunologic evaluation. Nontypeable H. influenzae lacks a polysac- have a severe allergic reaction to dry natural rubber latex. The vial
charide capsule that renders the Hib vaccine ineffective against this stoppers for Pentacel and MenHibRix do not contain latex.
cause of acute otitis media (AOM), acute sinusitis, bronchitis, and As with all pertussis-containing vaccines, benefits and risk should
community-acquired pneumonia. be considered before administering Pentacel to those with a history of
fever higher than 40.5°C, hypotonic-hyporesponsive episode, persistent
Adverse Events inconsolable crying lasting 3 or more hours within 48 hours after receipt
Hib vaccines are well tolerated. Local reactions occur in approximately of a pertussis-containing vaccine, or seizures within 3 days of receiving
25% of recipients but typically are mild and last less than 24 hours.232,680 a pertussis-containing vaccine.
Systemic reactions such as fever and irritability are infrequent occurrences. Hib monovalent and combination conjugate vaccines are inactivated
When conjugate vaccines are administered concurrently with DTaP vaccines and can be administered to people with immunocompromising
vaccine, the incidence of systemic reactions is similar to that observed conditions. However, immunologic response to the vaccine may be
when only DTaP is given.232 suboptimal.408

Indications Hepatitis A Vaccine


Routine vaccination against Hib disease is recommended for all children The occurrence of hepatitis A is highest in developing countries and
beginning at approximately 2 months of age.32,89 Two or three doses, reflects the primary route of transmission: fecal-oral, person-to-person
depending on the product, given at 2-month intervals (optimal) are spread. In the United States, before vaccine became available, hepatitis
indicated by the time that the child is 6 months of age. PedvaxHIB is A was a common infection that caused substantial morbidity with
given as a two-dose primary series at 2 and 4 months, whereas ActHIB, significant associated costs.151,400 In 1995, the hepatitis A vaccine was
Hiberix, Pentacel, and MenHibRix are approved for immunization at licensed by the FDA, and during the decades that followed, acute hepatitis
2, 4, 6, and 15 to 18 months of age (see Table 245.7). The recommended A infection in the United States dramatically declined, from 31,032
age to initiate the primary series is 2 months, with a minimum age of reported infections in 1995 to an all-time low of 1398 in 2011.270,494 In
6 weeks. The minimum interval between doses is 4 weeks, with at least 2012 and 2013, however, the first increases in cases occurred since 1995,
8 weeks separating the last dose in the primary series and the booster. with 1562 and 1781 reported cases, respectively.494
Excellent immune responses have been achieved when vaccines from In the prevaccine era, the incidence of hepatitis A varied considerably
different manufacturers have been interchanged in the primary among different populations.270,619 A shift occurred in the epidemiology
series.53,77,312 PRP-T and PRP-OMP are considered interchangeable for of hepatitis A in the United States after the introduction of hepatitis A
primary and booster vaccinations. If more than a single brand of vaccine vaccine. Disease in the United States most commonly occurred in children
is used, the child should receive a three-dose primary series. A final 5 to 14 years of age.270 After vaccines were licensed, rates of infection
dose of any product, irrespective of the previous vaccines received, is among children declined more rapidly than did rates among adults,
acceptable when the child is 12 to 15 months of age for completion of resulting in similar rates among all age groups. Historically, rates of
the Hib vaccine immunization schedule. When feasible, the conjugate infection were highest among Alaska Natives and American Indians,
vaccine product used for the first dose should be used for subsequent and most cases occurred in a small number of states and counties in
doses in children younger than 12 months. the western and southwestern regions of the country. During the decades
For American Indian and Alaska Natives, PRP-OMP is the preferred after vaccine licensure, we have seen near-elimination of age-specific,
vaccine for the primary series doses. Hib meningitis incidence peaks racial, and geographic disparities.270,494
at a younger age among American Indian and Alaska Native infants.218,608 Individuals at risk for hepatitis A infections include close contacts
PRP-OMP vaccine produces a protective antibody response after the of people infected with hepatitis A virus (HAV), travelers to developing
first dose and provides early protection that American Indian and Alaska countries, those who engage in homosexual and bisexual activity, and
Native infants particularly need.20,232,230 injecting drug users. However, in approximately 50% of reported cases,
For children in whom immunization for Hib infection has not been no risk factor is identified.33 These infections likely are attributable to
initiated by the time that they reach 7 months of age, the recommended fecal-oral spread from asymptomatic contacts. Because children frequently
schedules depend on the child’s age and the choice of conjugate have asymptomatic infections and can shed virus for prolonged periods,
vaccine.32,89 Previously unimmunized children between 15 and 59 months they play an important role in transmission of HAV. Children and
2554 PART V  Prevention of Infectious Diseases

infants shed virus for longer periods than adults do, up to several months hepatitis B vaccine (Twinrix) is licensed in the United States for people
after the onset of clinical illness. In one study involving adults without 18 years and older. All HAV-containing vaccines are administered
an identified source of infection, 52% of their households included a intramuscularly.
child younger than 6 years.270
It has long been recognized that the control and ultimate elimination Immunogenicity and Efficacy
of hepatitis A by active immunization could best be achieved through Inactivated hepatitis A vaccine is highly immunogenic. After receiving
universal childhood immunization.651 However, it was not until 2005 a single dose, 95% of children and most adults seroconvert within 1
that hepatitis A vaccine was licensed for use in children from 12 to 23 month.33,270 After receipt of a second dose in children, seroconversion
months of age. Before then, it had been restricted to children older approximates 100%. Hepatitis A vaccine is immunogenic in children
than 2 years and consequently could not be incorporated readily into younger than 2 years old who do not have passively acquired maternal
routine infant vaccinations. In May 2006, the ACIP recommended antibody.536,643 In two large clinical trials of inactivated hepatitis A vaccine
including hepatitis A vaccine in the routine infant immunization in children older than 2 years, the protective efficacy rate was greater
schedule.270 than 90%.358,682 In a double-blind, placebo-controlled, randomized study
Before vaccine was available, community-wide outbreaks, recurring in Thailand involving approximately 34,000 vaccinees, the protective
every 3 to 10 years in high-risk communities, accounted for much of efficacy rate against clinical hepatitis A was 94% after administration
the occurrence of hepatitis A infection and disease. Outbreaks among of two doses given 1 month apart; it was 100% after subsequent
children attending childcare and the staff are common occurrences and administration of a 12-month booster dose.358
have been associated with community outbreaks.326 However, the Vaccination has been effective in controlling outbreaks in communities
prevalence of hepatitis A infection among childcare center staff and with high rates of disease.270 For example, in a New York State community
the children and adolescents who previously attended is not increased, in which HAV was highly endemic among children, a single dose of
a finding suggesting that infections within childcare settings most vaccine was 100% effective beginning 3 weeks after immunization in
commonly reflect transmission within the community that extends to preventing symptomatic disease.682 Moreover, observations from countries
these settings.326 Transmission of HAV also can occur in institutions where routine hepatitis A vaccination of infants or children has been
for the developmentally disabled and in neonatal intensive care units. implemented suggest a strong herd immunity effect.221,240 The duration
Transmissions from hospitalized patients to health care professionals of protection after vaccination is likely to be prolonged. Protective
have been reported.33 antibody levels of anti-HAV were observed in 99% of children evaluated
In addition to direct person-to-person transmission, infection can 5 to 6 years after receiving Vaqta.681 Kinetic models of antibody decline
be acquired by the ingestion of contaminated water or food, especially indicate that protective levels of antibody could exist for more than 25
that imported from endemic countries. An outbreak in 2013 from years in adults and up to 20 years in children.650,652 No data are available
contaminated pomegranate seeds, which were imported to the United to determine whether and when children will need a hepatitis A booster
States from Turkey, infected 165 people across 10 states; 42% were vaccine. In an ongoing study designed to address the need for a booster
hospitalized, three developed fulminate hepatitis, and one required liver dose, no cases of hepatitis A among children 9 years after initial vaccina-
transplantation.494 tion have been reported.683
Reduced vaccine immunogenicity has been observed in infants with
Preparations passively acquired anti–hepatitis A antibody who are administered
Inactivated and attenuated HAV vaccines have been developed.239 hepatitis A vaccines.221,428,433 Studies demonstrated that despite lower
However, only inactivated vaccines are licensed in the United States. antibody levels in infants born to anti–hepatitis A–positive mothers,
Inactivated HAV vaccine is prepared by methods similar to those used most infants with passively acquired antibody had an anamnestic response
for inactivated poliomyelitis vaccine. Virus is propagated in human to a booster dose 1 to 6 years later.221,269,380 In most infants, maternally
diploid fibroblast cell cultures, formalin inactivated, and adsorbed to acquired anti–hepatitis A antibody declines to undetectable levels by
aluminum hydroxide adjuvant. Two inactivated HAV vaccines are licensed the time the child reaches 12 months of age.432 Hepatitis A vaccine is
in the United States: Havrix (SmithKline Beecham Biologicals) and highly immunogenic for all infants when administered when they are
Vaqta (Merck & Co.). Vaqta and Havrix have two formulations, an adult older than 1 year, irrespective of maternal antibody status.72,221
and a pediatric product with different antigen content (Table 245.8). Concurrent administration of immune globulin and vaccine inhibits
The pediatric formulation is indicated for people 12 months to 18 years the peak serum antibody concentration achieved but not the rate of
of age. The vaccines can be used interchangeably.95 seroconversion.666 Because antibody concentrations reach much higher
Limited data indicate that hepatitis A vaccine may be administered than protective levels, this inhibition is not considered clinically significant
simultaneously with other vaccinations.33 A combination HAV and and supports passive-active immunoprophylaxis when indicated.

TABLE 245.8  Recommended Doses and Schedules for Inactivated Hepatitis A Vaccines
Vaccine Hepatitis A
Age (y) Trade Name Antigen Dose Volume per Dose (mL) No. of Doses Schedule
1–18 Havrix 720 ELU 0.5 2 Initial and 6≥12 mo later
1–18 Vaqta 25 Ua 0.5 2 Initial and 6≥18 mo later
≥19 Havrix 1440 ELU 1.0 2 Initial and 6≥12 mo later
≥19 Vaqta 50 Ub 1.0 2 Initial and 6≥18 mo later
≥18 Twinrixc 720 ELU 1.0 3 or 4 Initial, 1 and 6 mo later
or
Initial, 7 days and 21–30 d, followed
by a dose at 12 mo
a
Antigen units; each unit is equivalent to 1 µg of viral protein.
b
A combination of hepatitis B (Engerix-B, 20 µg) and hepatitis A (Havrix, 720 ELU) vaccine.
c
Twinrix is licensed for use in people ≥18 years in 3-dose and 4-dose schedules.
ELU, Enzyme-linked immunosorbent assay units.
Modified from American Academy of Pediatrics. Hepatitis A. In: Kimberlin DW, Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious Diseases. 30th
ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:391–9.
CHAPTER 245  Active Immunizing Agents 2555

Adverse Events • Adolescent and adult users of injection and noninjection drugs
Except for rare reports of anaphylaxis and anaphylactoid reaction in should be vaccinated. Periodic outbreaks among injection and
adults in Europe and Asia, serious reactions to inactivated HAV vaccine noninjection drug users have been reported in many parts of the
have not been reported.269,380 Pain, tenderness, and infection at the United States and in Europe.
injection site can occur.358 • Susceptible patients with chronic clotting disorders who receive
clotting factor concentrates should be immunized. Reported outbreaks
Indications of hepatitis A in patients with hemophilia receiving solvent-detergent–
HAV immunization is recommended routinely for children 12 through treated factor VIII and factor IX concentrates were identified during
23 months of age, for people who are at increased risk for infection, for the 1990s, primarily in Europe, although one case was reported in
those who are at increased risk for severe manifestations of hepatitis A the United States.
if infected, and for anyone who wants to obtain immunity.22,33,151,156,270 • People at risk for occupational exposure (e.g., handlers of HAV-
All children in the United States should receive hepatitis A vaccine infected primates, people working with HAV in a research laboratory
routinely according to the licensed two-dose schedule, with the initial setting) should be immunized. Outbreaks of hepatitis A have been
dose administered at 1 year of age (i.e., 12 to 23 months) and the second reported among people working with nonhuman primates that were
dose 6 to 12 months later. Children who are not immunized by 2 years born in the wild rather than those born and raised in captivity.
of age can be immunized at subsequent visits. • People with chronic liver disease are at increased risk for fulminant
Hepatitis A vaccination is recommended routinely for the following hepatitis A. Susceptible patients who are awaiting or have received
people who are at increased risk for infection22,33,270: liver transplants should be immunized.
• All susceptible people traveling to or working in countries that have Although the ACIP guidelines do not recommend routine vaccination
a high or intermediate hepatitis A endemicity should be vaccinated of food handlers, vaccination of these workers should be considered
or receive immune globulin before departure.151 HAV vaccine at the in areas where state and local health authorities or private employers
age-appropriate dose is preferred to immune globulin. The first dose determine that vaccination is cost-effective.
of HAV vaccine should be administered as soon as travel is considered.
One dose of single-antigen vaccine administered at any time before Postexposure Prophylaxis
departure can provide adequate protection for most healthy people. People who have been exposed to HAV but have not received HAV
Travelers who are younger than 12 months should receive a single vaccine should have one dose of single-antigen HAV vaccine or immune
dose of immune globulin (0.02 mL/kg), which provides effective globulin as soon as possible.151 Table 245.9 provides prophylaxis guidance
protection for up to 3 months; 0.06 mL/kg for travel provides protec- and dosages. The efficacy of immune globulin or vaccine for postexposure
tion up to 6 months. prophylaxis when administered more than 2 weeks after exposure has
• Susceptible household members and other close personal contacts not been established. Information about the relative efficacy of vaccine
of international adoptees newly arriving from countries with high compared with immune globulin after exposure is limited, and no data
or intermediate hepatitis A endemicity should be vaccinated.27,156 are available for people older than 40 years or those with underlying
Data from a study conducted at three adoption clinics in the United medical conditions.
States indicate that 1% to 6% of newly arrived international adoptees All previously unimmunized people with close personal contact
have acute HAV infection. The risk of HAV infection among close with a person with serologically confirmed HAV infection, such as
personal contacts of international adoptees is estimated at 106 (range, household and sexual contacts, should receive vaccine or immune
90–819) per 100,000 household contacts of international adoptees globulin within 2 weeks after the most recent exposure. Serologic testing
within the first 60 days after their arrival in the United States. HAV of contacts is not recommended because testing adds unnecessary cost
vaccine should be administered to all previously unvaccinated people and may delay administration of postexposure prophylaxis.
who anticipate close personal contact (e.g., household contact, regular Outbreaks of HAV infection at childcare centers have been recognized
babysitters) with an international adoptee from a country with high since the 1970s, but their frequency has decreased as HAV immunization
or intermediate endemicity during the first 60 days after arrival of rates for children have increased and as hepatitis A incidence among
the adoptee in the United States. The first dose of the two-dose HAV children has declined. HAV vaccine or immune globulin should be
vaccine series should be administered as soon as adoption is planned, administered to all previously unimmunized staff members and attendees
ideally two or more weeks before the arrival of the adoptee. of childcare centers or homes if one or more cases of hepatitis A are
• Male adolescents and adults who have sex with men should be recognized in children or staff members or cases are recognized in two
immunized. Outbreaks of hepatitis A among men who have sex or more households of center attendees. In centers that provide care
with men have been reported often, including in urban areas in the only to children who do not wear diapers, vaccine or immune globulin
United States, Canada, and Australia. need be given only to classroom contacts of an index-case patient.

TABLE 245.9  Recommendations for Postexposure Immunoprophylaxis of Hepatitis A Virus


Time Since Exposure Age of Patient Recommended Prophylaxisa
≤2 wk <12 mo IGIM, 0.02 mL/kg
12 mo–40 y HAV vaccine
≥41 y IGIM, 0.02 mL/kg but HAV vaccine can be used if IGIM
is unavailable
People of any age who are immunocompromised, have chronic IGIM, 0.02 mL/kg
liver disease, or a contraindication to vaccination
≥2 wk <12 mo No prophylaxis
≥12 mo No prophylaxis, but HAV vaccine may be indicated for
ongoing exposure
a
Dosage and schedule of HAV vaccine as recommended according to age in Table 245.8.
HAV, Hepatitis A virus; IGIM, immune globulin intramuscular.
Modified from American Academy of Pediatrics. Hepatitis A. In: Kimberlin DW, Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious Diseases. 30th
ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015: 391–399.
2556 PART V  Prevention of Infectious Diseases

When an outbreak occurs (i.e., hepatitis A cases identified in two elements of this strategy included preventing perinatal transmission of
or more families), vaccine or immune globulin also should be considered HBV by identifying and providing immunoprophylaxis to infants of
for members of households that have children (i.e., center attendees) hepatitis B virus surface antigen (HbsAg)–positive mothers and universal
in diapers. Children and adults with hepatitis A should be excluded hepatitis B vaccination of infants to interrupt transmission and prevent
from the center until 1 week after onset of illness, until the postexposure future infection. This approach provided effective programs for routine
prophylaxis program has been completed in the center, or until directed childhood immunization, protection without any need to identify specific
by the health department. Although precise data concerning the risk factors, and protection before significant exposure occurred. The
onset of protection after postexposure prophylaxis are not available, positive effects of universal infant immunization had been observed in
allowing prophylaxis recipients to return to the childcare center setting Taiwan, where the strategy of universal infant immunization already
immediately after receipt of the vaccine or immune globulin dose seems was being used. In Taiwan, the overall prevalence rate of HBV for children
reasonable. 1 to 10 years of age decreased from 9.8% in 1984 to 1.3% in 1994.182
Schoolroom exposure usually does not pose an appreciable risk for In 1994, the ACIP expanded the recommendations to include previ-
infection, and postexposure prophylaxis is not indicated when a single ously unvaccinated children 11 to 12 years old.124 In October 1997,
case occurs and the source of infection is outside the school. However, these recommendations were expanded to include all unvaccinated
HAV vaccine or immune globulin can be used for unimmunized people children from birth to 18 years old and made hepatitis B vaccine available
who have close contact with the index patient if transmission within through the Vaccines for Children (VFC) program for those from birth
the school setting is documented. to 18 years of age who were eligible for the program.135 The goal of the
Usually, health care–associated HAV in hospital personnel has 1997 recommendations was to increase access to hepatitis B vaccine by
occurred through spread from patients with acute HAV infection for encouraging the vaccination of previously unvaccinated children and
whom the diagnosis was not recognized. Careful hygienic practices adolescents when they were seen for routine medical visits.15 Expansion
should be emphasized when a patient with jaundice or known or of the recommended age group for receiving vaccination and for VFC
suspected hepatitis A is admitted to the hospital. When outbreaks occur, eligibility simplified previous recommendations and eligibility criteria
HAV vaccine or immune globulin is recommended for people in close for receiving HBV vaccine. The other ACIP priorities for giving hepatitis
contact with infected patients. There is no recommendation for routine B vaccination to children remained unchanged and included the following
preexposure use of HAV vaccine for hospital personnel. groups: all infants; children in populations at high risk for HBV infection,
If a food handler is diagnosed with hepatitis A, HAV vaccine or such as Alaska Natives, Pacific Islanders, and children who reside in
immune globulin should be provided to other food handlers at the households of first-generation immigrants from countries where HBV
same establishment. Food handlers with acute HAV infection should infection is moderately or highly endemic; previously unvaccinated
be excluded for 1 week after the onset of illness. Because common-source children 11 to 12 years of age; and older adolescents and adults in
transmission to patrons is unlikely, postexposure prophylaxis with HAV defined at-risk groups.124,116
vaccine or immune globulin typically is not indicated but may be Between 1990 and 2004, the incidence of HBV infection in the United
considered if the food handler directly handled food during the time States declined by 75%. The decline was greatest among children and
when he or she likely was infectious and had diarrhea or poor hygiene adolescents. Coincident with the decline in HBV incidence, vaccine
practices and if prophylaxis can be provided within 2 weeks of exposure. coverage for children 19 to 35 months of age increased from 16% to
Routine HAV immunization of food handlers is not recommended. 92% from 1991 to 2004.147 Among adolescents 13 to 15 years old, vaccine
coverage increased from 0% to 74% between 1993 and 2004 according
Precautions and Contraindications to CDC data. Since its inception in 1991, many aspects of the national
Hepatitis A vaccine should not be administered to people with a immunization strategy to eliminate HBV transmission have been
hypersensitivity reaction to any of the vaccine components, such as implemented with great success, especially the routine vaccination of
alum or, in the case of Havrix, phenoxyethanol.15 Safety data for pregnant all infants. Nonetheless, many challenges remain.
women are not available, but the risk is considered to be low or nonexistent Despite the overall decline in hepatitis B incidence and the recom-
because the vaccine contains inactivated, purified viral proteins. mendations for routine vaccination of infants and children, increasing
proportions of new HBV infections in the United States occur among
Hepatitis B Vaccine adolescents and adults who have defined risk factors, including those
Hepatitis B virus (HBV) infection is a leading cause of acute hepatitis with multiple sex partners (i.e., more than one partner during the
and a major public health problem of global importance. Its incidence preceding 6 months), men who have sex with men, and injecting
is especially high in many Asian and African countries. Individuals with drug users.441 The primary means of preventing these infections is to
chronic infection are at risk for chronic hepatitis, cirrhosis, and primary identify settings such as correctional facilities, sexually transmitted
hepatocellular carcinoma. Rates of new infection are highest among disease clinics, and drug treatment centers where adolescents and adults
adults, but chronic infection is more likely to occur in individuals infected with high-risk drug and sexual practices can be routinely accessed and
as infants or children. They are at increased risk for chronic and malignant vaccinated. In correctional facilities where previously unvaccinated
liver disease, and as chronic carriers, they serve as the reservoir for inmates are offered HBV vaccination, 60% to 80% of inmates accepted
transmission of HBV. offered vaccination.677
The initial strategies for prevention of hepatitis B through vaccination Although universal screening of all pregnant women has been widely
reflect the various epidemiologic patterns of HBV infection in different adopted across the United States, fewer than half of all HBsAg-positive
areas of the world.116 In the United States, for example, infection is of pregnant women are identified through prenatal screening.261 These
comparatively low endemicity and occurs primarily in adolescents and women with unknown HBsAg status are not screened consistently, even
adults. The risk of infection, however, is much greater in certain popula- when they are hospitalized during labor and delivery; consequently,
tions. Examples include those born and living in areas or among groups their infants do not receive appropriate postexposure prophylaxis.635
in which HBV is highly endemic and those with lifestyles predisposing The birth dose of HBV vaccine, which can serve as a safety net for
to the acquisition of HBV, such as male homosexual activity, intravenous infants born to mothers for whom testing was not performed or not
drug abuse, and promiscuous heterosexual activity.11 In geographic areas performed correctly, was administered to only 45% of infants born in
in which HBV infection is highly endemic, infection usually is acquired the United States in 2004, less than the rate of 54% seen before July of
at birth or during childhood, a pattern prompting the recommendation 1999, when recommendations were made to suspend the birth dose of
for universal vaccination of infants. Many countries have adopted hepatitis hepatitis B vaccine temporarily until a thimerosal-free vaccine became
B vaccine into their national campaigns, and children recently immigrated available.440
to the United States from endemic areas confirm this trend, with higher To enhance existing strategies aimed at prevention of perinatal HBV
levels of neutralizing antibodies found in newer immigrants.709 transmission, the ACIP recommended in December of 2005 that all
In 1991, the CDC initiated a comprehensive hepatitis B vaccination delivery hospitals institute specific policies and procedures to improve
strategy to eliminate transmission of HBV in the United States.116 Critical identification of infants born to HBsAg-positive mothers and mothers
CHAPTER 245  Active Immunizing Agents 2557

with unknown HBsAg status.464 These recommendations were made Adverse Events
to ensure administration of appropriate postexposure prophylaxis and Other than soreness at the injection site, reactions to hepatitis B vaccine
a birth dose of HBV vaccine to all medically stable infants. A study of rarely occur. Postvaccination surveillance performed after licensure of
prospectively collected data from 5 of 64 US-funded Perinatal Hepatitis the plasma-derived vaccine indicated a possible association between
B Prevention Programs from 2007 through 2013 found that perinatal Guillain-Barré syndrome and receipt of the first vaccine dose, but no
hepatitis B virus infection occurred among 1.1% of infants, despite evidence indicates an association of Guillain-Barré syndrome with
94.9% of infants receiving hepatitis B vaccine and hepatitis B immune recombinant vaccine.464,465 Anaphylaxis has been estimated to occur in
globulin within 12 hours of birth.583 Infants born to mothers who were one of 600,000 doses distributed.464,465 Several nonfatal pediatric cases
younger, hepatitis B e antigen (HbeAg) positive, or who had a high have been reported.
viral load or received fewer than three hepatitis B vaccine doses were
at greatest risk for infection. Indications
The 2005 ACIP statement also updated recommendations to improve Routine immunization with hepatitis B vaccine is recommended for
vaccination coverage of adolescents and children by implementing all infants in the United States and should be completed by 6 to 18
immunization record reviews for all children 11 to 12 years old and months of age for all infants born to HBsAg-negative mothers.34,464
children younger than 19 years who were born in countries with high Delivery hospitals should develop policies that ensure administration
or intermediate HBV endemicity and by vaccinating all unvaccinated of a birth dose for all infants weighing 2000 g or more at birth
adolescents in settings that provide health care services to this age unless a physician’s order to defer immunization is in place and the
group.464 serologic status of the mother is in the infant’s medical record. Admin-
istering the first dose of hepatitis B vaccine soon after birth
Preparations should minimize the risk of infection because of errors in maternal
Hepatitis B vaccines consisting of inactivated, purified HBsAg derived HBsAg testing or reporting or from exposure to people with chronic
from chronic hepatitis B plasma were introduced in the early 1980s. In HBV infection in the household and can increase the likelihood of
the late 1980s, two recombinant vaccines (Recombivax HB, Merck & completing the vaccine series. For infants weighing less than 2 kg who
Co.; Engerix-B, SmithKline Beecham Biologicals) were licensed in the are born to HBsAg-negative mothers, initiation of vaccine should begin
United States and are available in single-antigen and combined formula- at 1 month of chronologic age. Administration of the second dose of
tions. Only the recombinant vaccines are available in the United States, vaccine is recommended 1 to 2 months after administration of the
but plasma-derived vaccines are widely used in other areas of the world. first dose, followed by a third dose when the infant is 6 to 18 months
The recombinant vaccines contain 5 to 40 µg/mL of HBsAg protein. of age.
Pediatric formulations contain no thimerosal or only trace amounts. Only single-antigen hepatitis B vaccine can be used for doses
Vaccine is administered intramuscularly in the anterolateral thigh given to infants between birth and 6 weeks of age. Single-antigen or
or deltoid area, depending on the age and size of the recipient. Admin- combination vaccine may be used to complete the series; four doses
istration in the buttocks or intradermally has been associated with of vaccine can be administered if a birth dose is given and a combination
decreased immunogenicity and is not recommended at any age. vaccine containing a hepatitis B component is used to complete
the series.
Immunogenicity and Efficacy Routine screening of all pregnant women for HBsAg is recommended
The recommended series of three doses of vaccine induces a protective because of the necessity for administering a birth dose of vaccine and
antibody response in more than 95% of infants, children, adolescents, HBIG.34,464 Infants born to HBsAg-positive mothers, including preterm
and adults younger than 40 years.34,464 In field trials, the efficacy and low-birth-weight infants, should be given HBIG (0.5 mL) within
rates have been 80% to 95% and usually correlate with immunogenicity. 12 hours after birth at a separate injection site, as well as the birth dose
Protection against disease is virtually 100% for people who develop of HBV vaccine.34,464 If a preterm infant weighing less than 2000 g is
adequate serum antibody concentrations (anti-HBs ≥10 mIU/mL) born to an HBsAg-positive mother, the birth dose of hepatitis B vaccine
after receiving vaccination. Adults older than 40 years and immunosup- should not be counted toward completion of the vaccine series, and
pressed people are less likely to develop protective anti-HBs three additional doses of hepatitis B vaccine should be administered
concentrations. beginning when the child is 1 month of age. In addition to receiving
Hepatitis B vaccine is highly immunogenic. Postvaccination serologic HBIG and the hepatitis B vaccine series within 12 hours of birth, infants
testing is not indicated except for infants born to HBsAg-positive women, of HBsAg-positive mothers should be tested for HBsAg and antibody
people with ongoing occupational exposure to blood, and people with to HBsAg (i.e., anti-HBs) at 9 to 12 months of age (or 1–2 months
immunosuppressive conditions. Active immunization in combination after the final dose of the vaccine series, if the series is delayed) to
with passive immunoprophylaxis with hepatitis B immune globulin identify those with chronic HBV infection and those who may require
(HBIG) administered within 12 to 24 hours after birth to infants born revaccination.464,582
to chronically infected mothers is more than 90% effective in preventing A woman whose HBsAg status is unknown at delivery should undergo
transmission of HBV to the infant. Active postexposure vaccination blood testing as soon as possible to determine it. The infant should
without HBIG administered soon after birth has been effective in prevent- receive the first dose of hepatitis B vaccine within 12 hours. If the
ing perinatal transmission and is used in areas where the use of HBIG woman is found to be HBsAg positive, her infant should receive HBIG
is impractical.310 as soon as possible within 7 days. In populations for which HBsAg
Vaccine-induced protection against symptomatic infection in a normal testing of pregnant women is not feasible, all infants should receive
host is prolonged and correlates with immunologic memory, which hepatitis B vaccine at birth and 2 months and complete the series by
has been demonstrated in immunized children and adults for at least the time they reach 6 months of age.
12 years after vaccination. Children immunized at birth are protected Hepatitis B vaccination is recommended for all children and ado-
for at least 10 years. The need for routine booster doses has not been lescents younger than 19 years in the United States. Children who
demonstrated. have not been immunized previously may begin the vaccine series
Vaccine failures related to HBV variants with mutations in the S during any visit. All children 11 to 12 years of age should have a
gene leading to conformational changes in the HBsAg protein, the major review of their immunization records and receive the complete
target for neutralizing anti-HBs antibody, have occurred in fully vac- hepatitis B vaccine series if they have not been vaccinated previously.
cinated children and perinatally exposed infants who received appropriate All children and adolescents younger than 19 years who were born
active and passive postexposure prophylaxis and in many cases had in Asia, the Pacific Islands, Africa, or other intermediate- or high-
protective anti-HBs antibody levels.356 Although no evidence establishes endemic countries or have at least one parent who was born in one of
that these escape mutations pose a threat to the effectiveness of current these areas should have a review of their immunization records and
hepatitis B vaccine programs, surveillance to detect emergence of HBV complete the hepatitis B vaccine series if they were not vaccinated
variants in immunized populations is warranted.464,717 previously.464
2558 PART V  Prevention of Infectious Diseases

TABLE 245.10  Recommended Doses and Schedules of Hepatitis B Virus Vaccines in the United States
VACCINE
Group Recombivax HB Dose µg (mL) Engerix-B Dose µg (mL)
Infants of HBsAg-negative mothers and children and adolescents <20 y 5 (0.5) 10 (0.5)
Infants of HBsAg-positive mothers (HBIG also recommended) 5 (0.5) 10 (0.5)
Children 1–10 y 5 (0.5)a 10 (0.5)a,b
Adolescents 11–15 y 10 (0.5)c N/A
Adolescents 11–19 y 5 (0.5)a 10 (0.5)a,b
Adults (≥20 y) 10 (1.0) 20 (1.0)b
Patients undergoing dialysis and other immunocompromised persons
  <20 yd 5 (0.5) 10 (0.5)
  ≥20 y 40 (1.0)e 40 (2.0)f
Unvaccinated adults with diabetes mellitus 19–59 yg 10 (1.0) 20 (1.0)
a
Pediatric formulation licensed for use in a 3-dose schedule at 0, 12, and 24 months for children 5 to 16 y and at 0, 1, and 6 mo for adolescents 11 to 16 y.
b
Also licensed as a 4-dose schedule at 0, 1, 2, and 12 mo for all age groups.
c
Adult formulation administered on a 2-dose schedule at 0 mo and then 4 to 6 mo later, licensed for adolescents 11 to 15 y.136
d
Higher doses may be more immunogenic, but no specific recommendations have been made.
e
Special formulation for patients undergoing dialysis given at 0, 1, and 6 mo.
f
Two 1.0-mL doses administered as one or two injections in a 4-dose schedule at 0, 1, 2, and 6 mo.
g
Three doses administered at 0, 1, and 6 months.172
HBIG, Hepatitis B immune globulin; HBsAg, hepatitis B surface antigen.
Modified from American Academy of Pediatrics. Hepatitis B. In: Kimberlin DW, Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious Diseases. 30th
ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:400–23; Centers for Disease Control and Prevention. A comprehensive immunization strategy to eliminate transmission of
hepatitis B virus infection in the United States: recommendations of the Advisory Committee on Immunization Practices (ACIP). Part 1: immunization of Infants, children, and adolescents.
MMWR Recomm Rep. 2005;54RR-16):1–32.

Vaccination also is recommended for those with one or more of the accident involving blood in a hospital or an injury to a susceptible
following risk factors for acquiring HBV infection34: person caused by the bite of an HBsAg-positive child. For this
• Sexually active adolescents and adults who have a recently acquired indication, HBIG and vaccine are given. Recommendations in these
sexually transmitted disease, are seeking evaluation or treatment for circumstances are complex and based on the availability of the blood
a sexually transmitted infection, are identified as sex workers, or source for HBsAg testing and the hepatitis B vaccination status of
have had one or more sex partners in the previous 6 months the exposed person.
• Sexually active men who have sex with men Booster doses are not recommended except for patients undergoing
• Household contacts or sexual partners of HBsAg-positive people hemodialysis and possibly other immunocompromised patients, who
• Injecting drug users should undergo annual antibody testing to assess the need. An additional
• People at occupational risk for infection through exposure to blood dose is indicated for those whose serum anti-HBsAg concentration is
or blood-contaminated body fluids, such as health care workers and less than 10 mIU/mL.
public safety workers The current recommended doses and schedule for the use of hepatitis
• Residents and staff of institutions for the developmentally disabled B vaccines licensed in the United States in infants and other age groups
• Patients undergoing hemodialysis (vaccination of those with early are shown in Table 245.10, and more information can be found else-
renal failure is encouraged before they require hemodialysis) where.34,464,465 For completing the hepatitis B vaccine series and achieving
• Patients with human immunodeficiency virus (HIV) infection, chronic complete vaccination for hepatitis B, the two licensed hepatitis B vaccines
liver disease, or diabetes mellitus are interchangeable when administered in doses recommended by the
• Members of households with international adoptees who are HBsAg manufacturers.34,464 In September 1999, the FDA approved an optional
positive two-dose schedule of Recombivax HB for vaccination of adolescents
• Travelers, especially children, to areas with high and intermediate 11 to 15 years of age. The ACIP recommended that this schedule be
rates of infection with HBV who have close contact with the local included in the VFC program in February 2000.136 With the two-dose
population or are likely to have contact with blood, such as in a schedule, the adult dose of Recombivax HB is administered to adolescents
medical setting or by sexual contact with residents 11 to 15 years old, with the second dose given 4 to 6 months after the
• Inmates in long-term correctional facilities first dose. In immunogenicity studies enrolling adolescents 11 to 15
In addition to active and passive immunoprophylaxis of infants years of age, antibody concentrations and seroprotection rates were
born to HBsAg-positive mothers, postexposure prophylaxis is recom- similar with the two-dose schedule and the currently licensed three-dose
mended in the following circumstances: schedule. Follow-up data collected during the course of 2 years indicated
• Sexual partner of an HBsAg-positive person. A single dose of HBIG that the rate of decline in concentration of antibody for the two-dose
within 14 days of the last sexual contact and initiation of the three- schedule was similar to that for the three-dose schedule.
dose hepatitis B vaccination is recommended for susceptible people. No data are available to assess long-term protection or immune memory
• Household exposure or close contact of an unvaccinated infant younger after vaccination with the two-dose schedule, and whether booster doses
than 12 months old to a primary caregiver who has acute or chronic of vaccine will be required is not known. Children and adolescents who
hepatitis B infection. Infants in this circumstance should receive HBIG have begun vaccination with a dose of the pediatric formulation should
and be vaccinated against hepatitis B. If at the time of exposure only complete the three-dose series with this dose. Similarly, if the formulation
one dose of vaccine has been administered, the second dose should administered to an adolescent at the start of a series is not known, the
be administered if the interval is appropriate, or HBIG should be series should be completed with the three-dose schedule.
administered if immunization is not yet due. If the infant has been A combination of hepatitis B (Engerix-B, 20 µg) and hepatitis A
immunized fully or has received at least two doses of vaccine, the (Havrix, 720 enzyme-linked immunosorbent assay units [ELU]) vaccine
infant should be presumed protected, and HBIG is not required. (i.e., Twinrix) is licensed for use in people 18 years or older in a three-dose
• Accidental percutaneous or per mucosal exposure of a susceptible schedule administered at birth, 1 month, and 6 or more months later.
person to HBsAg-positive blood, such as a needlestick or other An alternative four-dose schedule was approved in 2007; vaccinations
CHAPTER 245  Active Immunizing Agents 2559

are given at birth, day 7, and between days 21 and 30 and followed by Although the incidence of HPV is high, most infections clear without
a booster dose at 12 months.34 intervention. Seventy percent of new infections clear within 1 year, and
In December of 2006, the ACIP approved recommendations for 91% clear within 2 years.196 Persistent infection with high-risk HPV
hepatitis vaccine use in adults.465 In settings in which high proportions types, defined as detection of the same viral type at two or more visits
of adults are at risk for HBV infection, the ACIP recommends universal 6 months apart, is the most important risk factor for developing cervical
hepatitis B vaccination for all unvaccinated adults. Settings include cancer precursor lesions. Studies have demonstrated that persistent
sexually transmitted infection treatment facilities, HIV testing and infection with a high-risk HPV type is associated with a greater than
treatment facilities, facilities providing drug abuse treatment and preven- 10-fold risk of high-grade cervical cancer precursors.350,483
tion services, health care settings targeting services to injection drug Three prophylactic HPV subunit vaccines are licensed and recom-
users, correctional facilities, health care settings serving men who have mended for use in the United States.457,533 Use of these vaccines has
sex with men, chronic hemodialysis facilities and end-stage renal disease significantly decreased the prevalence of vaccine-type HPV infection.
programs, and institutions and nonresidential care facilities for people In one study, vaccine-type HPV prevalence in the study population
with developmental disabilities. decreased by 75% among women and more than 90% among vaccinated
For other primary care and specialty medical settings in which adults women.379 Areas with high HPV vaccine coverage have seen decreases
at risk for HBV infection receive care, the ACIP recommends that health in anogenital warts.244,245,547 The impact on the incidence of HPV-related
care providers inform all patients about the health benefits of vaccination, cervical and other anogenital cancers will take years to assess because
including the risk of acquiring HBV infection, and other people for the cancers can occur decades after infection. Incidence of recurrent
whom vaccination is recommended. They should vaccinate adults who respiratory papillomatosis and associated cancers also is likely to decrease
report a risk of developing HBV infection and any adults requesting as a result of widespread vaccination.
protection from HBV infection. To promote vaccination in all settings,
health care providers should implement standing orders to identify Preparations
adults recommended for hepatitis B vaccination and administer vac- In 2006, the FDA licensed the first HPV vaccine for use in girls and
cination as part of routine clinical services, not require acknowledgment women 9 through 26 years of age in the United States, and this was
of an HBV infection risk factor for adults to receive vaccine, and use done for use in boys and men 9 through 26 years of age in 2009.35,167,456
available reimbursement mechanisms to remove financial barriers to The quadrivalent vaccine (Gardasil, Merck & Co.) contains HPV types
receiving hepatitis B vaccination. 6, 11, 16, and 18 virus-like particles (VLPs) and protects against four
In 2011, the ACIP recommended that all previously unvaccinated HPV types, which together cause 70% of cervical cancers (i.e., HPV
adults 19 through 59 years of age with type 1 or type 2 diabetes mellitus types 18 and 16) and 90% of genital warts (i.e., HPV types 6 and 11).
be vaccinated against hepatitis B as soon as possible after a diagnosis The quadrivalent HPV vaccine (HPV4) is prepared from the highly
of diabetes is made. Unimmunized adults with diabetes mellitus who purified VLPs of the major capsid (L1) protein of HPV types 6, 11, 16,
are 60 years or older may be vaccinated at the discretion of the treating and 18. The L1 proteins are produced by Saccharomyces cerevisiae and
clinician after assessing their risk.172 self-assemble into VLPs. The VLPs mimic the HPV virus, but they
contain no viral DNA. Each 0.5-mL dose contains 20 µg of HPV 6 L1
Contraindications protein, 40 µg of HPV 11 L1 protein, 40 µg of HPV 16 L1 protein, and
The only contraindication to receiving HBV vaccination is a history of 20 µg of HPV 18 L1 protein.
anaphylaxis to a previous dose of vaccine. Data on the safety of HBV A bivalent vaccine (HPV2) (Cervarix, GlaxoSmithKline), which is
vaccines are not available for pregnant women, but because the vaccines directed against two oncogenic HPV types (i.e., 16 and 18), was licensed
contain only HBsAg and not live virus, they should not harm the for use in girls and women 10 through 25 years of age in 2009 and not
developing fetus. Because HBV infection during pregnancy can result licensed for males.35,159
in transmission to the neonate, susceptible women at increased risk for A 9-valent vaccine (9vHPV) (Gardasil 9, Merck), which protects
infection should be vaccinated during pregnancy. Inadvertent vaccination against the same four HPV types as the quadrivalent HPV vaccine plus
of HBsAg-positive people has no deleterious effects. HPV 31, 33, 45, 52, and 58 VPs, was approved by the FDA in 2014 and
recommended by the ACIP in 2015.277,533 It was licensed for use in men
Human Papillomavirus Vaccine and women 9 to 26 years of age.277
Genital HPV infection is thought to be the most common sexually The HPV vaccines use alum as an adjuvant and do not contain
transmitted viral infection, accounting for more than 14 million new thimerosal or antibiotics. The vaccines should be stored at 2°C to 8°C
cases annually in the United States.579 The estimated prevalence of HPV (36°F–46°F) and not frozen.159
infection among 15- to 19-year-olds is 32.9%, and among 20-to 24-year- Primary vaccination dosing schedules with HPV vaccines are the
old, sexually active women, it is 53.8%.579 Prevalence is thought to be same and consist of three 0.5-mL doses administered intramuscularly.
similar among males.249 HPV typically infects girls and women soon The second and third doses should be administered 2 and 6 months
after sexual exposure.208,690 Median time from first intercourse to first after administration of the first dose. In 2016, the ACIP recommended
detection of HPV is only 3 months.208 The estimated lifetime risk of that 9- to 14-year-old children receive two HPV vaccine doses instead
acquiring HPV infection by those with at least one sexual partner is of the previously recommended three doses because studies showed
more than 84% for women and more than 91% for men.189 More than good immune responses compared with earlier groups receiving three
80% of women and men acquire HPV infection by 45 years of age.189 doses.474 The recommendation for three doses remained the same for
More than 40 HPV types can infect the genital tract and are classified ages 15 to 26 years. The FDA also announced approval of the two-dose
on the basis of their association with cervical cancer.492 The low-risk, schedule for the 9vHPV vaccine.278 Due to licensing and manufacturing
nononcogenic HPV types (i.e., types 6 and 11) are associated with issues, 9vHPV will be the primary HPV vaccine going forward.
anogenital warts, mild cervical dysplasia, and recurrent respiratory
papillomatosis. The high-risk, oncogenic HPV types are causally linked Immunogenicity and Efficacy
to several human cancers in women and men, including cancers of the Data from randomized, controlled trials (RCTs) have shown consistently
cervix, vulva, vagina, anus, and penis and a subset of head and neck that prophylactic HPV VLP vaccines are immunogenic and efficacious
cancers.439 Infection with HPV causes almost all cases of cervical cancer, in preventing infection and lesions caused by the targeted HPV
and before vaccine development, it was the second most common cause types.336,402,439,476,616 Vaccine efficacy seems to be mediated by a type-specific
of death from cancer among women globally, surpassed only by breast humoral immune response.
cancer.439 Cervical cancer remains a significant cause of cancer death Evaluations of the quadrivalent HPV4 vaccine have demonstrated
worldwide.265 The development of cervical cancer often occurs decades that vaccinated individuals develop high antibody titers to the respective
after initial infection. HPV types 16 and 18 account for 70% of cervical HPV types that exceed the antibody titers seen with natural HPV
cancers.196 A variety of other types account for the remaining 30% of infection.616 However, the level of antibody that confers a protective
cervical cancers, and these types vary in their distribution globally. response is unknown.616 For the HPV4 VLP vaccine, an antibody response
2560 PART V  Prevention of Infectious Diseases

significantly greater than placebo was sustained for at least 3 years vaccinated previously or who have not completed the full vaccination
without booster doses.336,663 Although vaccine efficacy studies have been series.
done only enrolling adolescents 13 years or older, immunogenicity Only HPV4 and 9vHPV vaccines are recommended for routine
bridging studies have been performed with children as young as 9 years immunization of boys at 11 or 12 years of age (the series can be started
of age. Antibody titers to HPV types 6, 11, 16, and 18 were found to at 9 years of age), and it is recommended for boys and men 13 through
be higher in younger adolescents than in young adults. From these data, 21 years of age not previously immunized.209 Men 22 through 26 years
investigators inferred that protection exists against cervical cancer, cancer of age may be immunized on request. Vaccine is recommended for men
precursor lesions, and genital warts, even for young adolescent girls who have sex with men and males who are immunocompromised
who receive the HPV4 vaccine. (including those with HIV infection) through 26 years of age.
The results from four RCTs demonstrate that a regimen of three Ideally, vaccination should be administered before potential exposure
intramuscular injections of HPV VLP vaccine provides high-level to HPV through sexual contact occurs. HPV vaccines are not licensed
protection from infection and lesions caused by targeted HPV types.402,439 for use in people older than 26 years.35
A combined analysis of HPV4 vaccine efficacy in phase II and phase Vaccine is administered in a three-dose schedule, and the second
III clinical trials showed 100% protection against precancerous lesions and third doses should be administered 2 and 6 months after the first
(i.e., carcinoma in situ II/III or adenoma in situ) caused by HPV type dose. Immunogenicity data allow for some degree of flexibility in the
16 or 18 and 99% protection against genital wart development.476 dosing schedule, which is often an important consideration in providing
A randomized, multicenter, double-blind study found the 9vHPV multidose vaccines to adolescents for whom contact with health providers
vaccine antibody response to HPV types 6, 11, 16, and 18 to be non- can be sporadic. For both vaccines, the minimum interval from dose
inferior to the response with the HPV4 vaccine.378 From this result, the 1 to dose 2 is 4 weeks. The minimum interval from dose 2 to dose 3
9vHPV vaccine can be inferred to have the same protective efficacy as should not be less than 12 weeks. For ages 9 to 14 years, vaccine can
the HPV4 vaccine. The 9vHPV vaccine prevented infection and disease be administered in a two-dose schedule, with the second dose admin-
related to HPV types 31, 33, 45, 52, and 58; for high-grade cervical, istered at least 6 months after the first dose. The three-dose schedule
vulvar, or vaginal disease related to these HPV types, the vaccine had should be used for ages 15 to 26 years.474
a 96.7% efficacy rate.378 HPV vaccine can be administered at the same visit when other
It is not known whether HPV vaccines provide long-term protection age-appropriate vaccines, such as Tdap, Td, and MCV4 (meningococcal),
or booster doses will be necessary. The quadrivalent HPV4 vaccine does are provided.508 Women who have received an HPV vaccine must continue
not have efficacy against existing disease or infection caused by HPV. to have regular Papanicolaou test screening. HPV vaccines do not provide
HPV2 vaccine efficacy was evaluated in two randomized, double-blind, protection against all HPV types associated with development of cancer,
controlled clinical trials in females 15 through 25 years of age. In the and vaccines do not alter the progression of HPV infection acquired
analysis, 99% or more of participants developed an HPV 16 and 18 before immunization.
antibody response 1 month after completing the three-dose series. After People through 26 years of age with evidence of current HPV infec-
concomitant administration of HPV2 with tetanus toxoid, diphtheria tion, such as cervical dysplasia, a positive HPV DNA test result, or
toxoid, and acellular pertussis vaccine and/or with meningococcal anogenital warts or a history of anogenital warts, should receive HPV
conjugate vaccine in females 11 through 18 years of age compared with immunization because infection with all vaccine HPV types is unlikely
those after administration at separate visits, the antibody responses for and the vaccine can provide protection against HPV infection with
bivalent vaccine antigens were noninferior.159 types not already acquired.35 Vaccine recipients should be advised
In summary, HPV vaccines are highly effective in preventing HPV-16 that data from clinical trials do not indicate the vaccine will have any
and HPV-18–related cervical precancerous lesions in females. The HPV4 therapeutic effect on existing abnormalities found by Papanicolaou
vaccine is highly effective in preventing HPV-6 and HPV-11–related testing, on HPV infection, or on genital warts. However, HPV vaccination
genital warts in females and males, and HPV4 vaccine also is effective can provide protection against infection with HPV types not already
in preventing anal precancerous lesions in males. Vaccines offer no acquired.
protection against progression of infection to disease from HPV acquired Lactating women can receive HPV4 or 9vHPV vaccine, but the
before immunization.35 bivalent vaccine has not been studied in lactating women. Patients of
the appropriate age who are immunocompromised due to disease or
Adverse Events medication can receive HPV vaccine. However, the immune response
HPV vaccines appear to be generally safe and well tolerated.620 The to vaccination and vaccine effectiveness may be less than in those who
HPV4 and 9vHPV vaccines have similar safety profiles.533 The most are immunocompetent.
common adverse events are local injection site reactions such as mild
to moderate pain, redness, pruritus, or swelling.533 For females 9 to 26 Contraindications and Precautions
years of age, 9vHPV compared with HPV4 vaccine recipients had more HPV4 and 9vHPV vaccines are contraindicated for people with a history
injection site adverse events, including swelling (40.3% vs. 29.1%) and of immediate hypersensitivity to yeast or to any vaccine component. 533
redness (34.0% vs. 25.8%).533 Bivalent HPV2 vaccine is contraindicated for people with a history of
In analyses of local and general adverse events after bivalent HPV2 immediate hypersensitivity to any vaccine component. HPV2 vaccine
vaccine administration versus control vaccines, 92% reported injection in the prefilled syringe formulation should not be administered to
site pain, 48%, had redness, and 44% had swelling in the HPV2 group. latex-sensitive individuals because the rubber stopper contains latex.35
These findings compared with 64% to 87%, 24% to 28%, and 17% to HPV vaccine is listed under pregnancy category B and is not recom-
21% in the control groups for the same findings. Fatigue, headache, mended for use in pregnancy.159,457 If an adolescent becomes pregnant
and myalgia were the most common general symptoms reported.159 during the vaccination period, subsequent doses should be deferred
Additional data on vaccine safety, including data on pregnancy and until after parturition. The vaccine has not been associated causally
fetal and infant outcomes, are being collected. Data will continue to be with adverse outcomes of pregnancy or adverse effects on the developing
collected in postlicensure studies. fetus. However, data on vaccination during pregnancy are limited.

Indications Influenza Vaccine


The ACIP issued recommendations for the use of the HPV4 vaccine Influenza virus infection continues to cause significant morbidity and
in March 2007, the bivalent HPV2 vaccine in May 2010, and the mortality despite the availability of effective vaccines and antiviral therapy
9vHPV in March 2015.159,160,167,457,456,533 Routine vaccination with three for prevention and treatment of influenza. In the United States, epidemics
doses of vaccine is recommended for girls 11 to 12 years of age. The of influenza typically occur during the winter months and have been
vaccination series can be started in girls as young as 9 years of age at associated with an average of approximately 36,000 deaths per year,
the discretion of physicians and parents. Catch-up vaccination is recom- and between 1976 and 2007, estimated cases ranged from 3000 to 49,000
mended for girls and women 13 to 26 years of age who have not been each year.158,638
CHAPTER 245  Active Immunizing Agents 2561

Influenza viruses cause disease among all age groups.300,301,486 Rates the device included in the vaccine package. Vaccine is supplied in a
of infection are highest among children, but rates of serious illness and single-dose, prefilled microinjection system (0.1 mL). The intradermal
death are highest among those 65 years or older, children 2 years or formulation of IIV4 is shipped and stored at 2°C to 8°C (35°F–46°F).
younger, and people of any age who have medical conditions that increase Many manufacturers produce inactivated influenza vaccine each
the risk of complications from influenza.65,64,299,486 The impact of influenza year for the US market. Vaccines are available in single-dose syringes
on normal children and those with underlying high-risk conditions is and vials or multidose vials and in preservative-free formulations.
appreciable, and the burden of disease in young children attributed to Approved age indications vary by manufacturer and product. Clinicians
influenza is underrecognized.542 Attack rates among normal children should obtain inactivated influenza vaccine appropriate for the age
have been estimated at 10% to 40% each year, and approximately 1% groups they plan to vaccinate. The ACIP does not recommend use of
of influenza infections result in hospitalization.302,542 Influenza also causes influenza vaccine outside the vaccine’s FDA-approved age indication.
a significant burden in outpatient settings, especially for those 2 to 17 Tables listing each year’s influenza vaccines are available in the annual
years of age.279 ACIP and AAP influenza statements and on the CDC influenza website
A major difficulty in the development and provision of satisfactory (https://www.cdc.gov/flu/professionals/vaccination/vaccine_safety.htm).
immunizing agents for the prevention of influenza is the antigenic Package inserts should be consulted for recommended age groups and
variation of the viruses. Periodic minor antigenic changes in influenza possible contraindications for each vaccine in addition to information
A or B virus are the major factors in the continuing occurrence of regarding additional components of various vaccine formulations.
yearly influenza. Although outbreaks usually are limited in magnitude, LAIV is cold adapted, developed by passaging the viruses at succes-
the resulting morbidity and mortality remain discouragingly high. Major sively lower temperatures in tissue culture so that replication occurs
antigenic changes in influenza A virus, as occurred in 1957 to 1958 only in the upper respiratory tract.74 LAIV is licensed by the FDA for
(Asian strain), in 1968 to 1969 (Hong Kong strain), and again in 2009 healthy individuals 2 through 49 years of age. It has not been recom-
(pH1N1) account for the pandemic spread of disease associated with mended for those with a history of asthma or other high-risk medical
greater overall morbidity and mortality. The CDC estimates that in the conditions associated with an increased risk of complications from
2009 pandemic the H1N1 influenza virus caused more than 60 million influenza.
Americans to become ill and led to more than 270,000 hospitalizations LAIV is administered intranasally in a prefilled, single-use sprayer
and 12,500 deaths.371 People younger than 65 years accounted for 90% containing 0.2 mL of vaccine. A removable dose-divider clip is attached
of hospitalizations and deaths. With typical seasonal influenza, approxi- to the sprayer to administer 0.1 mL separately into each nostril. Children
mately 90% of the people who die are older than 65 years. younger than 9 years being immunized against influenza for the first
time should receive two doses of LAIV given 4 weeks apart before the
Preparations start of the influenza season. The LAIV formulation licensed in the
An assortment of influenza vaccines are licensed for use in the United United States must be shipped and stored at 2°C to 8°C (35°F–46°F).
States: inactivated trivalent influenza vaccine (IIV3), adjuvanted After the vaccine is warmed to room temperature for intended use, it
inactivated trivalent influenza vaccine (aIIV3), recombinant trivalent must be used within 30 minutes.
influenza vaccine (RIV3), inactivated quadrivalent influenza vaccine Trivalent influenza vaccines contain A (H1N1), A (H3N2), and B
(IIV4), inactivated quadrivalent cell culture-based influenza vaccine viral antigens. The quadrivalent influenza vaccines include an additional
(ccIIV4), and live attenuated quadrivalent influenza vaccine (LAIV).101,315 B viral antigen because of the difficulty of predicting which B virus
The trivalent vaccines contain three virus strains (usually two type lineage will predominate during a given season.
A and one type B), and the composition is changed periodically in
anticipation of the prevalent influenza strains expected to circulate in Immunogenicity and Efficacy
the United States in the following winter. The quadrivalent vaccines Children 6 months or older typically have protective levels of anti-
include an additional type B strain.315 Most vaccines are prepared from influenza antibody against specific influenza virus strains after receiving
virus grown in the allantoic sac of the chick embryo. Exceptions include the recommended number of doses of seasonal inactivated influenza
one IIV4 preparation with virus propagated through canine kidney vaccine.226,305,417,499,702,704 Immunogenicity studies using the influenza A
cells and the RIV3 preparation, which is manufactured without the use (H1N1) 2009 monovalent vaccine indicated that more than 90% of
of influenza viruses.315 children 9 years or older responded to a single dose with anti-influenza
IIV3, aIIV3, IIV4, and ccIIV4 are inactivated influenza vaccines (IIVs) antibody levels considered to be protective. Young children had incon-
that contain no live virus. IIVs distributed in the United States consist sistent responses to a single dose of the influenza A (H1N1) 2009
of subvirion vaccine, prepared by disrupting the lipid-containing monovalent vaccine across studies, with 20% of children 6 to 35 months
membrane of the virus, or purified surface-antigen vaccine. All IIV of age responding to a single dose with protective anti-influenza antibody
formulations are available for intramuscular use, except ccIIV4, which levels. However, in all studies, 80% to 95% of vaccinated infants, children,
is intradermal. and adolescents developed protective anti-influenza antibody levels to
IIV4 is licensed and recommended for children 6 months or older the 2009 H1N1 influenza virus after two doses.56,506,538
and for adults, including those with or without chronic medical condi- In most seasons, one or more seasonal vaccine antigens are changed
tions. Vaccine is available in pediatric (0.25-mL dose) and adult (0.5-mL compared with the previous season. In consecutive years when vaccine
dose) formulations. IIV3 and ccIIV4 (using the 0.5-mL dose) are licensed antigens change, children younger than 9 years who received only one
and recommended for children 4 years or older and for adults. RIV3, dose of vaccine in their first year of vaccination are less likely to have
also in the 0.5-mL dose, is licensed and recommended for those18 years protective antibody responses when administered only a single dose
or older. during their second year of vaccination compared with children who
IIV is administered intramuscularly into the anterolateral thigh of received two doses in their first year of vaccination.258,502,668
infants and young children and into the deltoid muscle of older children When the vaccine antigens do not change from one season to the
and adults. RIV3 is administered intramuscularly into the deltoid muscle. next, priming children 6 to 23 months of age with a single dose of
The volume of vaccine depends on age. Infants and toddlers 6 months vaccine in the spring followed by a dose in the fall results in similar
through 35 months of age should receive a dose of 0.25 mL, and all antibody responses compared with a regimen of two doses in the fall.257
individuals 36 months or older should receive 0.5 mL per dose. Children However, one study conducted during a season when the vaccine antigens
younger than 9 years who are being immunized against influenza for did not change compared with the previous season estimated 62%
the first time should receive two doses of TIV given 4 weeks apart before effectiveness against influenza-like illness for healthy children who had
the start of the influenza season. IIV is shipped and stored at 2°C to received only one dose in the previous influenza season and only one
8°C (35°F–46°F). dose in the study season compared with 82% for those who received
The intradermal formulation of IIV4 is licensed for the 2016–2017 two doses separated by 4 weeks or more during the study season.10
influenza season for use in individuals 18 through 64 years of age.315 The antibody response for children at higher risk for influenza-related
It is administered preferably over the deltoid muscle and only by using complications (e.g., children with chronic medical conditions) may be
2562 PART V  Prevention of Infectious Diseases

lower than those reported typically for healthy children.71,316 However, laboratory-confirmed influenza type B infection, respectively. Vaccinated
antibody responses among children with asthma are similar to those children 2 to 6 years of age with asthma did not have substantially fewer
of healthy children and are not substantially altered during asthma type B influenza virus infections compared with the control group in
exacerbations requiring short-term prednisone treatment.522 this study.623
Vaccine effectiveness studies also have indicated that two doses are The association between vaccination and prevention of asthma
needed to provide adequate protection during the first season that exacerbations is unclear. Vaccination provided protection against asthma
young children are vaccinated. Among children younger than 5 years exacerbations in some studies.403,511
who have never received influenza vaccine previously or who received IIV has reduced AOM in some studies. Two studies reported that
only one dose of influenza vaccine in their first year of vaccination, IIV decreased the risk of influenza-related otitis media by approximately
vaccine effectiveness is lower compared with children who received two 30% among children with mean ages of 20 and 27 months, respec-
doses in their first year of being vaccinated. Two large, retrospective tively.194,339 However, a large study conducted among children with a
studies of young children who had received only one dose of IIV in mean age of 14 months indicated that IIV was not effective against
their first year of being vaccinated found no decrease in influenza-like AOM.351 Influenza vaccine effectiveness against a nonspecific clinical
illness–related office visits compared with unvaccinated children.10,558 outcome such as AOM, which is caused by a variety of pathogens and
Similar results were reported in a case-control study of children 6 to is not typically diagnosed using influenza virus culture, is expected to
59 months of age in which laboratory-confirmed influenza was the be relatively low.
outcome measured.602 These results, along with the immunogenicity The relative effectiveness of LAIV and IIV for medically attended,
data indicating that antibody responses are substantially higher when laboratory-confirmed influenza was evaluated using US Influenza Vaccine
young children are given two doses, are the basis for the recommendation Effectiveness Network data.192 For the 2013–14 season, children 2 to 8
that all children 6 months to 8 years of age who are being vaccinated years of age who received LAIV had significantly higher odds of influenza
for the first time should receive two vaccine doses separated by 4 weeks (odds ratio, 5.36) than those who received IIV192 The greater odds of
or more. illness with influenza A/H1N1pdm09 included the 2010–11 and 2013–14
Estimates of vaccine efficacy or effectiveness among children 6 months seasons. Another study of the 2013–14 season found the effectiveness
or older have varied by season and study design. In a randomized trial of LAIV against influenza A/(H1N1)pdm09 to be 17%, compared with
conducted during five influenza seasons (1985–90) in the United States 60% for IIV, for children 2 to 17 years of age.284
for children 1 to 15 years of age, annual vaccination reduced laboratory- After the ACIP recommendation for preferential use of LAIV during
confirmed influenza A substantially (77–91%).501 A 1-year, placebo- the 2014–15 season for children 2 to 8 years of age, another study examined
controlled study reported vaccine efficacy rates against laboratory-confirmed vaccine effectiveness by type.716 In this study with 9311 participants at
influenza illness of 56% for healthy children 3 to 9 years of age and 5 US sites, the preference for use of LAIV in young children was not
100% for healthy children and adolescents 10 to 18 years old.197 supported. Based on these and several other studies, LAIV was not recom-
A randomized, double-blind, placebo-controlled trial conducted mended for use for any age for the 2016–17 season.101,315
during two influenza seasons among children 6 to 24 months of age
indicated that the efficacy rate was 66% against culture-confirmed Adverse Events
influenza illness during the 1999–2000 influenza season but did not All inactivated influenza vaccines contain killed viruses and therefore
reduce culture-confirmed influenza illness substantially during the cannot produce signs or symptoms of influenza caused by active virus
2000–01 influenza season.351 A case-control study conducted during infection. The most common symptoms associated with IIV administra-
the 2003–04 season indicated vaccine effectiveness of 49% against tion are soreness at the injection site and fever. Fever, usually occurring
laboratory-confirmed influenza.602 An observational study of children 6 to 24 hours after immunization, affects 10% to 35% of children younger
6 to 59 months of age with laboratory-confirmed influenza compared than 2 years. Mild systemic symptoms, such as nausea, lethargy, headache,
with children who tested negative for influenza reported vaccine muscle aches, and chills also can occur after receipt of IIV.
effectiveness of 44% in the 2003–04 influenza season and 57% during Before the 2010–11 influenza season, an increased risk of febrile
the 2004–05 season.255 Partial vaccination (i.e., only one dose for children seizures after IIV3 administration had not been observed in the United
being vaccinated for the first time) was not effective in either study. States.313,329 The CDC and the FDA conducted enhanced monitoring
During an influenza season (2003–04) with a suboptimal vaccine for febrile seizures after influenza vaccination because of reports of an
match, a retrospective cohort study conducted among approximately increased risk of fever and febrile seizures in young children in Australia
30,000 children 6 months to 8 years of age indicated vaccine effectiveness that were associated with a 2010 Southern Hemisphere vaccine produced
of 51% against medically attended, clinically diagnosed pneumonia or by CSL Biotherapies. The risk was up to nine febrile seizures per 1000
influenza (i.e., no laboratory confirmation of influenza) among fully doses during the 2010–11 influenza season. Due to the increased risk,
vaccinated children and 49% among approximately 5000 children 6 to the ACIP does not recommend the US licensed Afluria (CSL Biotherapies’
23 months old.558 IIV3) for children younger than 9 years.164
Another retrospective cohort study of similar size conducted during For the 2010–11 season, surveillance for US licensed influenza vaccines
the same influenza season in Denver but limited to healthy children 6 detected safety signals for febrile seizures in younger children after IIV
to 21 months of age estimated clinical effectiveness of 2 IIV doses to administration.426,645 On further review, the increased risk was found for
be 87% against pneumonia or influenza-related office visits.10 children 6 months through 4 years of age, ranging from the day of vac-
Among children, TIV effectiveness may increase with age.501,711 A cination to the day after vaccination (i.e., 0–1-day risk window). The
systematic review of published studies estimated vaccine effectiveness risk was found to be higher when children received concomitant PCV13,
at 59% for children 2 years or older but concluded that additional and the risk peaked at about 16 months of age.645 No increased risk was
evidence was needed to demonstrate effectiveness among children 6 observed for children 5 years or older after IIV or for children of any
months to 2 years old.370 age after LAIV. The magnitude of the increased risk of febrile seizures
Because of the recognized influenza-related disease burden among in young children in the United States (<1 case/1000 children vaccinated)
children with other chronic diseases or immunosuppression and the was substantially lower than the risk observed in Australia in 2010.
long-standing recommendation for vaccination of these children, After evaluating the data on febrile seizures from the 2010–11
randomized, placebo-controlled studies to determine efficacy in high-risk influenza season and taking into consideration benefits and risks of
children are needed but are currently unavailable. Although data about vaccination, no policy change was recommended for use of IIV or
influenza vaccine in specific immunocompromised states are limited, PCV13. According to the CDC, surveillance data on febrile seizures in
the Infectious Diseases Society of America (IDSA) publishes guidelines young children after administration of influenza vaccine for the 2011–12
for specific immune-compromising diagnoses.568 In a nonrandomized, influenza season (i.e., same vaccine formulation as for the 2010–11
controlled trial enrolling children 2 to 6 years old and 7 to 14 years old season) were consistent with those from the 2010–11 influenza season.
who had asthma, vaccine efficacy was 54% and 78% against laboratory- No changes in the use of IIV or PCV13 are recommended, although
confirmed influenza type A infection and 22% and 60% against surveillance is ongoing.315
CHAPTER 245  Active Immunizing Agents 2563

LAIV usually is well tolerated and may produce mild signs or


symptoms related to influenza virus infection that include headache Has the child received 2 total doses of
and runny nose or nasal congestion in vaccinees. Transmission of LAIV trivalent or quadrivalent influenza
strains to unimmunized contacts has been documented only once in vaccine before July 1, 2016? (doses need not
prelicensure studies. The proposed explanation for the uncommon have been received during the same
occurrence of transmission is that the vaccine virus is shed for a shorter season or consecutive seasons)
duration and in a much smaller quantity than are wild-type strains.

Indications
Routine annual influenza vaccination is recommended for all people
6 months or older.37,101,315 To permit time for production of protective
antibody levels,91,317 vaccination optimally should occur before the onset Yes No or don’t know
of influenza activity in the community. Vaccination providers should
offer vaccination as soon as the vaccine is available. Vaccination should
be offered throughout the influenza season (i.e., as long as influenza
viruses are circulating in the community).101,315 Due to the previously 1 dose of 2016–17 2 doses of 2016–17
described issues with vaccine effectiveness, LAIV was not recommended influenza vaccine influenza vaccine
by the ACIP for any age group for the 2016–17 influenza season.315 (administered
Particular focus should be on the administration of IIV for all children 4 weeks apart)
and adolescents who have underlying medical conditions associated
with an increased risk of complications from influenza, including the
FIG. 245.2  Influenza vaccine dosing algorithm for children age 6 months
following: through 8 years for the US 2016–17 influenza season. (From Centers
• Asthma or other chronic pulmonary diseases, including cystic for Disease Control and Prevention. Prevention and control of seasonal
fibrosis influenza with vaccines recommendations of the Advisory Committee
• Hemodynamically significant cardiac disease on Immunization Practices—United States, 2016–17 influenza season.
• Immunosuppressive disorders or therapy https://www.cdc.gov/mmwr/volumes/65/rr/rr6505a1.htm.)
• HIV infection
• Sickle cell anemia and other hemoglobinopathies
• Diseases that require long-term aspirin therapy, including juvenile year. The AAP recommends that two doses be given to these children
idiopathic arthritis or Kawasaki disease the following influenza season.101 This recommendation applies only
• Chronic renal dysfunction to the influenza season that follows the first year that a child younger
• Chronic metabolic disease, including diabetes mellitus than 9 years old receives influenza vaccine (Fig. 245.2).
• Any condition that can compromise respiratory function or handling
of secretions or can increase the risk of aspiration, such as neuro- Precautions and Contraindications
developmental disorders, spinal cord injuries, seizure disorders, or IIVs are contraindicated for infants younger than 6 months and children
neuromuscular abnormalities who have a moderate to severe febrile illness on the basis of clinical
Although universal immunization for all individuals 6 months or judgment of the pediatrician.179 People with acute febrile illness usually
older is recommended, particular immunization efforts should be made should not be vaccinated until their symptoms have abated. However,
for the following groups to prevent transmission of influenza to those minor illnesses with or without fever do not contraindicate the use of
at risk, unless contraindicated: influenza vaccine, particularly in children with mild upper respiratory
• Household contacts and out-of-home care providers of children tract infection or allergic rhinitis.101,315
younger than 5 years and at-risk children of all ages should be Pregnancy is not a contraindication to influenza vaccine administra-
immunized. tion. Vaccination with IIVs are advised for pregnant girls and women
• Any female who is pregnant, is considering pregnancy, has just who have an underlying high-risk condition.
delivered, or is breastfeeding during the influenza season should be IIVs can be used to prevent influenza in those who are in close
immunized.. Studies have shown that infants born to immunized contact with most immunosuppressed individuals, such as those receiving
women have better influenza-related health outcomes. However, the care in a protective environment after undergoing hematopoietic stem
estimated median seasonal vaccination coverage among women with cell transplantation.
a live birth was only 47% during the 2009–10 influenza season, even Severe allergic and anaphylactic reactions can occur in response to
though pregnant women and their infants are at higher risk for several influenza vaccine components, but such reactions are rare.
complications. Data from some studies suggest that influenza vac- Currently available influenza vaccines are prepared by means of inocula-
cination during pregnancy may decrease the risk of preterm birth. tion of virus into chicken eggs, with the exception of the RIV3 and
• Health care personnel or health care volunteers should be immunized.. ccIIV4 preparations.315 The use of influenza vaccines for people with
Despite the recent AAP recommendation for mandatory influenza a history of egg allergy was reviewed by the ACIP.315 For the 2016–17
immunization for all health care personnel,76,101 many remain unvac- influenza season, the ACIP recommended that people with egg allergy
cinated. The CDC estimated that only 77% of health care personnel who report only hives after egg exposure should receive IIVs, with
received the seasonal influenza vaccine for the 2014–15 season.81 several additional safety measures. For the 2016–17 influenza season,
The AAP recommends mandatory vaccination of health care personnel the ACIP recommended the following:
because they frequently come into contact in their clinical settings • People with a history of egg allergy who have experienced only hives
with patients at high risk for influenza illness. after exposure to egg should receive influenza vaccine.315
• Close contacts of immunosuppressed individuals should be • People who report having had reactions to egg involving symptoms
immunized. such as angioedema, respiratory distress, lightheadedness, or recurrent
Previously unimmunized children between 6 months to 9 years of emesis or who required epinephrine or another emergency medical
age should receive two doses of influenza vaccine before the onset of intervention, particularly those that occurred immediately or within
influenza season. Available data suggest that children younger than 9 minutes to hours after egg exposure, should receive influenza
years who did not receive the second dose of influenza vaccine in the vaccine.315
initial year that influenza vaccine was given may not be adequately
protected the next influenza season with only one dose. In this group, Measles Vaccine
levels of protection can be suboptimal, especially if the antigenic Since the introduction of an inactivated and a live virus, attenuated
specificity of the predominant strains has changed from the previous measles vaccine (i.e., Edmonston B strain) in the United States in 1963,
2564 PART V  Prevention of Infectious Diseases

the reported incidence of measles has decreased by more than 99%. (MMRV) vaccines. MMRV vaccine was licensed in the United States
Although the incidence of measles has declined in all age groups, the in September 2005 and may be used instead of MMR and varicella
decline has been greatest among children 5 to 14 years of age. vaccine to implement the recommended two-dose vaccine schedule for
Measles was targeted for elimination in the United States by 1982. prevention of measles, mumps, rubella, and varicella among children
Efforts to eliminate measles were not successful as a result of two factors. 12 months to 12 years of age. Single-component measles vaccine is not
The first was vaccine failure. In the late 1980s, outbreaks occurred among available in the United States.
older children in schools in which immunization rates usually were Inadequate protection against measles can result from the administra-
greater than 95%.459,505 Attack rates were 1% to 5% because of the tion of improperly stored vaccine. Before reconstitution, measles vaccine
accumulation of measles-susceptible individuals from vaccine failure. must be stored at a temperature between 2°C and 8°C (35.6°F–46.4°F)
The recurrent measles outbreaks among vaccinated school-aged children or colder and must be protected from light, which may inactivate the
in the mid-1980s prompted the ACIP and the AAP in 1989 to recommend virus. Reconstituted vaccine should be stored in a refrigerator and
that all children be given two doses of measles-containing vaccine, discarded if not used within 8 hours.
preferably as MMR.13,109 Although administration of the second dose
originally was recommended at entry to primary school (ACIP) or Immunogenicity and Efficacy
middle/secondary school (AAP), the ACIP, the AAP, and the AAFP now Immunization produces a mild or unapparent, noncommunicable
recommend that a child be given the second dose at age 4 to 6 years infection. Measles antibodies develop in approximately 95% of children
rather than delaying it until the child is 11 to 12 years old.38,471 The vaccinated at 12 months of age and in 98% of children vaccinated at
major benefit of administering the second dose is a reduction in the 15 months of age.455 Studies indicate that serologic evidence of measles
proportion of people who remain susceptible because of primary vaccine immunity develops in more than 99% of people who receive two doses
failure. Waning immunity is not a major cause of vaccine failure and of measles vaccine, separated by at least 1 month, on or after their first
has little influence on transmission of measles, and revaccination of birthday.191,206 Although vaccine-induced antibody titers are lower than
children who have low concentrations of measles antibody produces those after natural disease, persistence of protective titers for as long
only a transient rise in antibody concentration.454,458,505,518,669 as 16 years after administration of vaccine has been demonstrated.410,458
The second factor leading to outbreaks of measles was the failure Most vaccinated people who appear to lose antibody have an anamnestic
to implement current immunization strategies, especially in the inner response after revaccination, indicating that they most likely are still
cities, where a high proportion of preschool-aged children had not immune.514 A small percentage of vaccinated individuals may lose
been vaccinated. From 1989 through 1991, the proportion of unvaccinated protection after several years as a result of secondary vaccine failure.466
people with measles increased, as reflected by outbreaks among unvac-
cinated inner-city preschool-aged children. Many barriers to providing Adverse Events
timely immunization to these children were identified during investiga- Vaccine-associated symptoms, consisting of fever higher than 39.4°C
tion of the measles resurgence that occurred between 1989 and 1991. (102.9°F) or rash occurring 5 to 10 days after immunization, develop
Reported cases of measles declined rapidly after the 1989–91 resurgence in 5% to 18% of recipients.421,530 Serious complications related to vaccine
because of intensive efforts to vaccinate preschool-aged children. Measles use occur far less frequently than after natural measles.525
vaccination levels among 2-year-old children increased from 70% in Thrombocytopenia occurs at a rate of one case for every 30,000 to
1990 to 91% in 1997. From 2001 to 2008, a median of 56 measles cases 40,000 doses distributed. Based on data from Sweden and Finland, the
were reported to the CDC annually.523 IOM concluded that a causal association exists between MMR and
In 2000, the United States achieved measles elimination, defined as thrombocytopenia.362 The decrease in platelet count presumably is caused
interruption of year-round endemic measles transmission.382 However, by the measles component and usually is not clinically apparent. However,
importations of measles into the United States continue to occur, posing thrombocytopenic purpura occurring after vaccination has been reported.
risks for measles outbreaks and sustained measles transmission. Since Central nervous system disease, specifically encephalitis or encepha-
2000, the annual number of cases has ranged from a low of 37 in 2004 lopathy, is reported at a rate of less than one case per 1 million doses
to a high of 667 in 2014.267,287 Most cases have been among people who of vaccine administered. Because the incidence of encephalitis or
are not vaccinated against measles. Most cases are imported from other encephalopathy after the administration of measles vaccination to healthy
countries or linked to imported cases. Most imported cases originate children is lower than the observed incidence of encephalitis of unknown
in Asia, Africa, and Europe and occur among US citizens traveling origin, some or most of the reported severe neurologic disorders may
abroad and people visiting the United States from other countries.614 be only temporally, rather than causally, related to measles immunization.
Since 1993, the largest outbreaks of measles in the United States have The risk of subacute sclerosing panencephalitis (SSPE) developing in
occurred in populations that refuse vaccination for religious or personal vaccinated children is extremely low and is estimated to be approximately
belief reasons.534,610 Most outbreaks have involved limited spread from one twelfth of the risk for SSPE developing after a case of natural
measles imported from outside the United States. The increased numbers measles (0.7 SSPE cases/1 million vaccine doses vs. 8.5 cases/1 million
of outbreaks and measles importations into the United States underscore natural measles infections).82,106,362 Vaccine-strain measles virus never
the ongoing risk of measles among unvaccinated people and the has been confirmed in a case of SSPE.38
importance of vaccination against measles. Reactions to measles vaccine are not age related and occur only in
The recommended age for receiving routine vaccination with measles susceptible vaccinees. After revaccination, reactions should be expected
vaccine has been lowered in the past decade from 15 months to 12 to 15 only in those who failed to respond to the first immunization.
months of age.141 The decision to lower the age for receiving routine No convincing evidence establishes that any vaccine causes autism
primary vaccination was based on the observation that most children or autistic spectrum disorder. Concern has been raised about a possible
are susceptible to measles by the time that they reach 12 months of age relation between MMR vaccine and autism by some parents of children
because of waning transplacental immunity.447,455 Most mothers have with autism. Symptoms of autism often are noticed by parents during
vaccine-induced immunity rather than immunity conferred by infection the second year of life and may manifest after administration of MMR
with wild virus. Because antibody concentrations induced by measles by weeks or months. Two independent nongovernmental groups, the
vaccination typically are lower than those induced by natural measles, IOM and the AAP, reviewed the evidence regarding a potential link
measles-specific antibodies acquired transplacentally are lower in infants between autism and MMR vaccine.328,360,361 Both groups independently
of vaccinated mothers, and these infants are susceptible at an earlier age. concluded that available evidence does not support an association and
that the United States should continue its current MMR vaccination
Preparations policy.
The live measles virus vaccine (Moraten strain) available in the United
States is prepared in chick fibroblast cell culture. Each dose of vaccine Indications
contains neomycin, sorbitol, and hydrolyzed gelatin as a stabilizer. Unless otherwise contraindicated, measles vaccine is indicated for people
Preparations include MMR and measles-mumps-rubella-varicella susceptible to measles.38,471 The recommended age for receiving the first
CHAPTER 245  Active Immunizing Agents 2565

dose of measles vaccine is 12 to 15 months. In high-risk areas, such as be administered for the first dose in this age group. For the second dose
those with recurrent measles transmission, the initial dose should be of measles, mumps, rubella, and varicella vaccines at any age (i.e., 15
administered when the child reaches 12 months of age. The second months to 12 years) and for the first dose at an age older than 48
dose is given routinely at 4 to 6 years and no later than 11 to 12 years months, use of MMRV vaccine usually is preferred over separate injections
of age. The minimal interval between the two doses is 4 weeks. of its equivalent component vaccines (i.e., MMR vaccine and varicella
Adults born before 1957 usually can be considered immune to measles vaccine). This recommendation is consistent with the ACIP’s 2011 general
because of previous natural infection. Those born after 1956 and for recommendations regarding use of combination vaccines, which state
whom immunoprophylaxis is indicated should receive two doses of that use of a combination vaccine usually is preferred over its equivalent
vaccine. component vaccines.408
During outbreaks, when the likelihood of exposure to measles is
high, measles vaccine should be given to infants as young as 6 months. Precautions and Contraindications
Seroconversion rates are significantly less for children vaccinated before Immunocompromised patients with conditions such as lymphoreticular
reaching 1 year of age than those for older children. Children immunized or other generalized malignant disease and primary or secondary
before their first birthday then should be revaccinated with MMR at immunodeficiency states should not be given live virus, attenuated
12 to 15 months of age, with a third dose given according to local policy. measles vaccine.38,471 After cessation of their chemotherapy, these individu-
Measles remains endemic in many areas of the world. Although als usually should not receive measles vaccine for at least 3 months.
vaccination against measles is not a requirement for entry into any However, because the intensity and type of immunosuppressive therapy,
country, susceptible children, adolescents, and adults born after 1956 radiation therapy, underlying disease, and other factors determine when
should be offered measles vaccination (usually as MMR) before embark- immunologic responsiveness will be restored, arriving at a definitive
ing on international travel. Infants 6 months or older who are traveling recommendation for an interval after cessation of immunosuppressive
to areas where measles is endemic or epidemic should be vaccinated therapy when measles vaccine can be safely and effectively administered
before departure and revaccinated at 12 to 15 months. Vaccination of often is not possible.
infants younger than 6 months is not necessary because most young An exception to the contraindication of administering measles vaccine
infants are protected by maternally derived antibodies. to immunocompromised patients is asymptomatic HIV-infected patients,
Exposure of susceptible individuals to measles is not a contraindica- for whom measles vaccination given as MMR at 12 to 15 months of
tion to administering vaccination; MMR vaccine given within 72 hours age is recommended. The need to protect HIV-infected people who are
of exposure may provide protection. For vaccine-eligible people 12 at increased risk for severe complications if infected with measles has
months or older who are exposed to measles, administration of MMR been balanced against the risk of adverse reactions. Measles vaccine is
vaccine is preferable to using immune globulin if administered within not recommended for HIV-infected people with evidence of severe
72 hours of initial exposure. If exposure does not result in infection, immunosuppression. A case of progressive measles pneumonitis occurred
immunization will protect against future infection. in a person with acquired immunodeficiency syndrome (AIDS) and
MMRV vaccine may be used instead of MMR and varicella vaccine severe immunosuppression to whom MMR vaccine was administered,126
to implement the recommended two-dose vaccine schedule for children and morbidity related to measles vaccination has been reported for
12 months to 12 years of age. At the time of its licensure, use of MMRV people with severe immunosuppression unrelated to HIV infection.
vaccine was preferred for the first and second doses over separate The antibody response to measles vaccine in severely immuno-
injections of equivalent component vaccines (i.e., MMR vaccine and compromised HIV-infected people is diminished.58
varicella vaccine), which was consistent with the ACIP’s 2006 general In the United States, the incidence of measles currently is very low.
recommendations on the use of combination vaccines.407 Among HIV-infected people who do not have evidence of severe
In February 2008, on the basis of preliminary data from two studies immunosuppression, no serious or unusual adverse events have been
conducted after licensure that suggested an increased risk of febrile reported after receiving measles vaccination.470,512,519,615 MMR vaccination
seizures 5 to 12 days after vaccination among children 12 to 23 months is recommended for all asymptomatic HIV-infected people who do not
of age who had received the first dose of MMRV vaccine compared have evidence of severe immunosuppression and for whom measles
with children the same age who had received the first dose of MMR vaccination would otherwise be indicated. MMR vaccination also should
vaccine and varicella vaccine administered as separate injections at the be considered for all symptomatic HIV-infected people who do not
same visit, the ACIP issued updated recommendations regarding MMRV have evidence of severe immunosuppression.568 Testing asymptomatic
vaccine use.155 The updated recommendations expressed no preference people for HIV is not necessary before administering MMR.17
for use of MMRV vaccine over separate injections of equivalent com- Systemically absorbed corticosteroids can suppress the immune
ponent vaccines for the first and second doses. system of an otherwise healthy person. However, neither the minimal
The final results of the two postlicensure studies indicated that among dose nor the duration of therapy sufficient to cause immune suppression
children 12 to 23 months of age, one additional febrile seizure occurred is well defined. Although the immunosuppressive effects of corticosteroid
5 to 12 days after vaccination per 2300 to 2600 children who had received treatment vary, many clinicians consider that a corticosteroid dose
the first dose of MMRV vaccine compared with children who had equivalent to or greater than a prednisone dose of 2 mg/kg per day or
received the first dose of MMR vaccine and varicella vaccine administered a total of 20 mg/day is sufficiently immunosuppressive to raise concern
as separate injections at the same visit.394 Data from postlicensure studies about the safety of administering live virus vaccines. People who have
do not suggest that children 4 to 6 years of age who received the second received systemic corticosteroids in doses of 2 mg/kg per day or 20 mg
dose of MMRV vaccine had an increased risk of febrile seizures after daily or on alternate days for an interval of 14 days or longer should
vaccination compared with children the same age who received MMR avoid receiving vaccination with MMR for at least 1 month after cessation
vaccine and varicella vaccine administered as separate injections at the of corticosteroid therapy.471,568 People who have received prolonged or
same visit.395 extensive topical, aerosolized, or other local corticosteroid therapy that
In June 2009, after consideration of the postlicensure data and other causes clinical or laboratory evidence of systemic immunosuppression
evidence, the ACIP adopted new recommendations regarding use of also should avoid receiving vaccination with MMR for at least 1 month
MMRV vaccine for the first and second doses and identified a personal after cessation of therapy.471
or family (i.e., sibling or parent) history of seizure as a precaution for The live attenuated measles virus vaccine used for immunization is
the use of MMRV vaccine.450 For the first dose of measles, mumps, not communicable. Contacts of immunocompromised patients
rubella, and varicella vaccines at 12 to 47 months of age, the MMR should be vaccinated to prevent the spread of natural measles to these
vaccine and varicella vaccine or MMRV vaccine may be used. Providers patients.
who are considering administering MMRV vaccine should discuss the Although no direct evidence has demonstrated that measles vaccine
benefits and risks of vaccination options with the parents or caregivers. is harmful to a pregnant woman or her fetus, the vaccine should not
Unless the parent or caregiver expresses a preference for MMRV vaccine, be administered to women known to be pregnant or who are considering
the CDC recommends that MMR vaccine and varicella vaccine should becoming pregnant because of the theoretical risk of fetal infection
2566 PART V  Prevention of Infectious Diseases

TABLE 245.11  Suggested Intervals Between Immune Globulin Administration and Measles or
Varicella Vaccination
DOSE
Product and Indication Route U or mL mg/kg Interval (mo)a
RSV monoclonal antibody (Synagis) IM — 15 (monoclonal) None
Tetanus (as TIG) IM 250 U 10 3
Hepatitis A prophylaxis (as IG)
  Contact prophylaxis IM 0.02 mL/kg 3.3 3
  International travel IM 0.06 mL/kg 10 3
Hepatitis B prophylaxis (as HBIG) IM 0.06 mL/kg 10 3
Rabies prophylaxis (as RIG) IM 20 IU/kg 22 4
Varicella prophylaxis (as VariZIG) IM 125 U/10 kg (maximum, 625 U) 20–40 5
Measles prophylaxis (as IG)
 Standard IM 0.25 mL/kg 40 5
  Immunocompromised contact IM 0.50 mL/kg 80 6
Botulinum immune globulin Intravenous (human, as BabyBIG) IV 1.5 mL/kg 75 6
Blood transfusion
  Washed RBCs IV 10 mL/kg Negligible None
  RBCs, adenine-saline added IV 10 mL/kg 10 3
  Packed RBCs IV 10 mL/kg 20–60 5
  Whole blood IV 10 mL/kg 80–100 6
  Plasma/platelet products IV 10 mL/kg 160 7
CMV immune globulin IV 3 mL/kg 150 6
IGIV
  Replacement therapy for immune deficiencies IV — 330–400 8
  Therapy for ITP IV — 400 8
  Varicella prophylaxis — 400 8
  Therapy for ITP IV — 1000 10
  Therapy for ITP or Kawasaki disease IV — 1600–2000 11
a
These intervals should provide sufficient time for decreases in passive antibodies in all children to allow an adequate response to measles or varicella vaccine. Physicians should not
assume that children are fully protected against measles during these intervals. Additional doses of immune globulin or measles vaccine may be indicated after exposure to measles.
BabyBIG, Botulinum immune globulin; CMV, cytomegalovirus; HBIG, hepatitis B immune globulin; IG, immune globulin; IM, intramuscular; ITP, immune thrombocytopenic purpura; IV,
intravenous; IGIV intravenous immune globulin; RIG, rabies immune globulin; RBCs, red blood cells; RSV, respiratory syncytial virus; TIG, tetanus immune globulin; VariZIG, varicella-zoster
immune globulin.
Modified from American Academy of Pediatrics. Active Immunization in People Who Recently Received Immune Globulin and Other Blood Products. In: Kimberlin DW, Brady MT, Jackson
MA, Long SS, editors. Red Book: 2015 Report of the Committee on Infectious Diseases. 30th ed. pp. 38-40, Elk Grove Village, IL: American Academy of Pediatrics; 2015 and Kroger A,
Sumaya C, Pickering L, et al. General recommendations on immunization—recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep.
2011;60:1-64.

associated with a live virus vaccine. Women vaccinated with MMR thrombocytopenia after receiving vaccination or from natural infection
should avoid conception for 28 days after being vaccinated.139 with measles or rubella. For children in whom thrombocytopenia
Because measles vaccination may diminish cutaneous manifestations develops in the month after receiving a dose of measles-containing
of cell-mediated immunity temporarily, a tuberculin test performed vaccine, withholding the second dose of measles vaccine is prudent if
several days to 6 weeks after receiving immunization can yield a false- the incidence of measles remains low.
negative result. Although natural measles infection can exacerbate Receipt of antibody-containing blood products (i.e., whole blood,
tuberculosis, no evidence indicates that measles vaccination is associated plasma, or parenteral immune globulin) may interfere with seroconver-
with such an effect. Tuberculin skin testing is not a prerequisite for sion in response to the measles vaccine. High doses of immune globulin
administering measles immunization. If a tuberculin test is indicated, preparations can inhibit the immune response to measles vaccine for
it should be performed on the day of immunization or postponed for 3 or more months, depending on the dosage.603 The length of time that
4 to 6 weeks because measles vaccination may suppress tuberculin passively acquired antibody persists depends on the concentration and
reactivity temporarily. quantity of the blood product received (Table 245.11).38,471
For people who are allergic to eggs, the risk of having serious allergic As with any condition that induces fever during the second year of
reactions such as anaphylaxis after receiving MMR vaccine is extremely life, children predisposed to having febrile seizures may experience
low, and skin testing with vaccine cannot predict an allergic reaction seizures after receiving measles vaccination. Most convulsions that
to vaccination.38,369,386 Obtaining a skin test is not required before develop after receiving measles immunization are simple febrile seizures
administering MMR to people who are allergic to eggs. Similarly, the and occur in children without known risk factors. Febrile seizures that
administration of gradually increasing doses of vaccine is not required.471 occur after the administration of vaccinations do not increase the risk
Data indicate that most anaphylactic reactions to measles- and mumps- of epilepsy or other neurologic disorders.66 The risk of seizures after
containing vaccines are not associated with hypersensitivity to egg receiving measles vaccination may be increased for children with a
antigens but rather to other components of the vaccines, such as the history of convulsions or those with a history of convulsions in first-
gelatin stabilizer.311,340,385,419 degree family members.662 Although the exact risk cannot be determined,
Children with a history of thrombocytopenic purpura or throm- it appears to be low. The recommendation to immunize children with
bocytopenia may be at risk for clinically significant thrombocytopenia a personal history of seizures or those with a history of seizures in
after receiving immunization with MMR.70,362 The decision to first-degree family members is based on factors indicating that the
vaccinate should be based on the benefits of immunity to measles, benefits greatly outweigh the risks. Prophylactic use of anticonvulsants
mumps, and rubella and the risk of recurrence or exacerbation of the usually is not feasible because therapeutic concentrations of many
CHAPTER 245  Active Immunizing Agents 2567

currently prescribed anticonvulsants are not achieved for some time 2009 through 2015, there were seven outbreaks of serogroup B meningitis
after the initiation of therapy. at US universities that resulted in 43 cases and 3 deaths.
In other parts of the world, the number of cases is much higher.368
Meningococcal Vaccine In the sub-Saharan African meningitis belt, which extends from Ethiopia
N. meningitidis causes a spectrum of infections, including meningitis, to Senegal, peaks of serogroup A meningococcal disease occur regularly
bacteremia, and rarely bacteremic pneumonia. Meningococcal disease during the dry season. Major epidemics occur every 8 to 12 years. In
develops rapidly and is fatal in 10% to 15% of cases. Of patients who each epidemic, tens of thousands of cases and thousands of deaths can
recover, 11% to 19% have permanent hearing loss, neurologic disability, occur. Approximately 350 million people are at risk. The phased introduc-
loss of limbs, or other serious sequelae. tion of MenAfriVac, a novel serogroup A meningococcal conjugate
In the United States since the introduction of Hib and pneumococcal vaccine that is being implemented through preventive national campaigns
polysaccharide-protein conjugate vaccines for infants, N meningitidis in meningitis belt countries for all individuals 1 to 29 years of age,
has become the leading cause of bacterial meningitis in children and holds great promise to end epidemic meningitis.406
remains an important cause of bacteremia and sepsis.586,686 Disease Meningococcal disease continues to cause epidemics outside the
most often occurs in previously healthy children 2 years or younger; meningitis belt.368 Serogroup W135 has emerged as an epidemic strain
the peak incidence occurs among children younger than 1 year.442 Another causing disease in Saudi Arabia in association with the Hajj pilgrimage.
peak occurs among adolescents and young adults 16 through 21 years Since 2002, serogroup W meningococcal disease has also been reported
of age. Historically, college freshman who lived in dormitories and in sub-Saharan African countries during epidemic seasons. More recently,
military recruits in boot camp had a higher rate of disease compared serogroup W outbreaks have occurred in South America. Serogroup X,
with people who are the same age and who are not living in such previously a rare cause of sporadic meningitis, has been responsible
accommodations. for outbreaks between 2006 and 2010 in Kenya, Niger, Togo, Uganda,
Close contacts of patients with meningococcal disease are at increased and Burkina Faso.705 Prolonged outbreaks of serogroup B meningococcal
risk for infection. The attack rate for household contacts is 500 to 800 disease have occurred in New Zealand, France, and Oregon. Serogroup
times the rate in the general population. Patients with persistent comple- Y emerged in Colombia and Venezuela, where it became the common
ment component deficiencies (i.e., C5 to C9, properdin, or factor H or disease-causing serogroup in 2006.571
factor D deficiencies) or anatomic or functional asplenia are at increased
risk for invasive and recurrent meningococcal disease. Although surveil- Preparations
lance data for cases of meningococcal disease among HIV-infected MenACWY vaccines.  Four meningococcal vaccines that contain purified
persons are limited in the United States, a growing body of evidence capsular polysaccharide alone or that are conjugated to a carrier protein
demonstrates an increased risk of meningococcal disease among HIV- are licensed and available in the United States for the prevention of
infected people.445 invasive disease caused by N. meningitidis serogroups A, C, W135, and
There are 12 known serogroups of N. meningitidis. Six serogroups Y (Table 245.12).
(i.e., A, B, C, W, X, and Y) are responsible for invasive human disease. Quadrivalent meningococcal polysaccharide vaccine (MPSV4;
Serogroups A and X are rare in the United States. Distribution of the Menomune, Sanofi Pasteur) was licensed in 1981 for use in children 2
other serogroups in the United States has shifted in the past 2 decades.205 years or older. Each vaccine dose contains 50 mg of each of the four
Serogroups B, C, and Y each account for approximately 30% of reported purified bacterial capsular polysaccharides from serogroups A, C, Y,
cases. Approximately three fourths of cases among adolescents and and W135. MPSV4 is available as a single dose or multidose (10-dose)
young adults are caused by serogroups C, Y, or W135. Among infants, vial of lyophilized vaccine, with a corresponding single-dose or multidose
50% to 60% of cases are caused by serogroup B. vial of diluent. After reconstitution of the lyophilized vaccine with the
For unknown reasons, the incidence of meningococcal disease has diluent, each dose consists of a 0.5-mL solution for injection. Vaccine
declined since the peak of disease in the late 1990s.205 Approximately supplied in single-dose vials should be used immediately after reconstitu-
800 to 1200 cases are reported annually in the United States. The decline tion. Vaccine supplied in multidose vials may be used for up to 35 days
began before the implementation of routine adolescent conjugate after reconstitution if stored at 2°C to 28°C (35°F–46°F). MPSV4 is
polysaccharide meningococcal immunization and has occurred in all administered subcutaneously and can be given concurrently with other
serogroups, including those not included in the available vaccines. vaccines but at different anatomic sites.
In the United States, the incidences of serogroups C and Y, which Two meningococcal conjugate vaccines (i.e., MenACWY-D [Menactra,
represent most cases of meningococcal disease, are at historic lows. Sanofi Pasteur] and MenACWY-CRM [Menveo, Novartis Vaccines]) are
However, a peak in disease incidence among adolescents and young licensed for use in people 2 through 55 years of age. MenACWY-D is a
adults 16 to 21 years of age has persisted, even after routine vaccination liquid solution supplied in 0.5-mL single-dose vials. Each dose contains
of adolescents was recommended in 2005. From 2006 through 2010, 4 µg each of the four capsular polysaccharides from serogroups A, C, Y,
the first 5 years after routine use of meningococcal vaccine was recom- and W135 conjugated to 48 µg of diphtheria toxoid. MenACWY-CRM
mended, the CDC received reports of approximately 30 cases of serogroup consists of two components: 10 µg of lyophilized meningococcal serogroup
C and Y meningococcal disease among people who had received the A capsular polysaccharide conjugated to CRM197 (MenA) and 5 µg each
vaccine. The case-fatality ratio was similar among people who had of capsular polysaccharide of serogroup C, Y, and W135 conjugated to
received vaccine compared with those who were unvaccinated. Of the CRM197 in 0.5 mL of phosphate buffered saline, which is used to recon-
13 reports of breakthrough disease for which data on underlying condi- stitute the lyophilized MenA component before injection. The reconstituted
tions were available, four people had underlying conditions or behaviors vaccine should be used immediately but may be held at or below 25°C
associated with an increased risk of bacterial infections, including type (77°F) for up to 8 hours. Meningococcal conjugate vaccines are administered
1 diabetes mellitus, current smoking, history of bacterial meningitis intramuscularly as a single 0.5-mL dose and can be given concurrently
and recurrent infections, and aplastic anemia, paroxysmal nocturnal with other recommended vaccines. Limited data suggest that different
hemoglobinuria, and receipt of eculizumab, an immune modulator conjugate vaccine products can be used interchangeably.204
that blocks complement protein C5.443 MenACWY-D and MenACWY-CRM are also licensed for infants
Serogroup B disease incidence has declined despite the fact that but with different administration schedules. MenACWY-D is licensed
serogroup B is not contained in any of the existing conjugate polysac- as a two-dose primary series, 3 months apart, for children 9 through
charide meningococcal vaccines. Approximately 50 to 60 cases of 23 months of age. MenACWY-CRM is licensed for use in children 2
serogroup B meningococcal disease occur annually among adolescents through 23 months of age. For children initiating vaccination at 2 months
and young adults 11 through 24 years of age in the United States.205 of age, MenACWY-CRM is administered as a four-dose series at 2, 4,
Outbreaks occur in communities and institutions, including childcare 6, and 12 months of age. For children initiating vaccination at 7 months
centers, schools, colleges, and military recruit camps. In the United through 23 months of age, MenACWY-CRM is administered as a two-
States, approximately 98% of cases of meningococcal disease are sporadic; dose series with the second dose administered in the second year of
however, outbreaks of meningococcal disease continue to occur.93 From life and at least 3 months after the first dose.
2568 PART V  Prevention of Infectious Diseases

TABLE 245.12  Licensed Meningococcal Vaccines—United States, 1981–2015


Formulation Type Trade Name Manufacturer Licensed (y) Age Group Doses Serogroups
MPSV4a
Polysaccharide Menomune Sanofi Pasteur 1981 ≥2 y Single dose A, C, W, and Y
MenACWY-Db Conjugate Menactra Sanofi Pasteur 2005 11–55 y Single dose A, C, W, and Y
MenACWY-Db Conjugate Menactra Sanofi Pasteur 2007 2–10 y Single dose A, C, W, and Y
MenACWY-Db Conjugate Menactra Sanofi Pasteur 2011 9–23 mo 2-dose series A, C, W, and Y
MenACWY-CRMc Conjugate Menveo Novartis 2010 11–55 y Single dose A, C, W, and Y
MenACWY-CRMc Conjugate Menveo Novartis 2011 2–10 y Single dose A, C, W, and Y
MenACWY-CRMc Conjugate Menveo Novartis 2011 2–23 mo 4-dose series A, C, W, and Y
Hib-MenCY-TTd Conjugate MenHibrix GlaxoSmithKline 2012 6 wk–18 mo 4-dose series C and Y
MenB-FHbpe Recombinant Trumenba Wyeth 2014 10–25 y 3-dose series B
MenB-FHbpe Recombinant Trumenba Wyeth 2014 10–25 y 2-dose series B
MenB-4Cf Recombinant Bexsero Novartis 2015 10–25 y 2-dose series B
a
Package insert available at http://www.fda.gov/downloads/BiologicsBloodVaccines/Vaccines/ApprovedProducts/UCM308370.pdf.
b
Package insert available at http://www.fda.gov/downloads/BiologicBloodVaccines/Vaccines/ApprovedProducts/UCM131170.pdf.
c
Package insert available at http://www.fda.gov/downloads/BiologicsBloodVaccines/Vaccines/ApprovedProducts/UCM201349.pdf.
d
Package insert available at http://www.fda.gov/downloads/BiologicsBloodVaccines/Vaccines/ApprovedProducts/UCM308577.pdf.
e
Package insert available at http://www.fda.gov/downloads/BiologicsBloodVaccines/Vaccines/ApprovedProducts/UCM421139.pdf.
f
Package insert available at http://www.fda.gov/downloads/BiologicsBloodVaccines/Vaccines/ApprovedProducts/UCM431447.pdf.

In June 2012, a combination conjugate vaccine against Hib (i.e., MenB-4C is licensed as a two-dose series, with doses administered at
PRP-T) and N. meningitidis serogroups C and Y (i.e., Hib-MenCY-TT least 1 month apart.
[MenHibrix, GlaxoSmithKline]) was licensed. Hib-MenCY-TT contains
5 µg of N. meningitidis serogroup C capsular polysaccharide, 5 µg of Immunogenicity and Efficacy
N. meningitidis serogroup Y capsular polysaccharide, and 2.5 µg of MenACWY vaccines.  The polysaccharide capsule of N meningitidis
Haemophilus influenzae serogroup B capsular polysaccharide, with each is an important determinant of virulence. Serum antibody to capsular
conjugated to tetanus toxoid. The vaccine is lyophilized and should be polysaccharide protects against disease by activating complement-
reconstituted with a 0.9% saline diluent . After reconstitution of the mediated bacteriolysis or opsonization, or both. Complement-dependent
lyophilized vaccine with the diluent, each dose consists of a 0.5-mL bactericidal activity induced by immunization with meningococcal
solution for injection. Hib-MenCY-TT is administered intramuscularly vaccine is measured by use of a serum bactericidal antibody assay
and is approved as a four-dose series for children at 2, 4, 6, and 12 with a human (hSBA) complement source. A defined bactericidal
through 18 months. antibody titer that indicates protection against invasive meningococcal
MenB vaccines.  In October 2014, the first serogroup B meningococcal disease is assay dependent. When sera are tested using a human
vaccine, MenB-FHbp (Trumenba, Wyeth Pharmaceuticals) was licensed. complement source, SBA titers of 1 : 4 or higher are considered
A second serogroup B vaccine, MenB-4C (Bexero, Novartis Vaccines), protective. Studies have demonstrated that almost all people who
was licensed in January 2015 (see Table 245.12). Both vaccines are developed invasive serogroup C meningococcal disease had sera that
licensed for use in people 10 through 25 years of age. lacked bactericidal activity to the pathogenic meningococcal strain.304,307
MenB-FHbp is a bivalent vaccine consisting of two recombinant In contrast, people with detectable SBA against a specific strain rarely
lipidated factor H binding protein (FHbp) antigens, one from developed disease.
FHbp subfamily A and one from subfamily B. FHbp is one of many The characteristics of MPSV4 are similar to other polysaccharide
proteins found on the surface of meningococci and contributes to the vaccines (e.g., pneumococcal polysaccharide). The vaccine is usually
ability of the bacterium to avoid host defenses. The FHbp proteins are not effective in children younger than 18 months. The response to the
individually produced in Escherichia coli. Each 0.5-mL dose contains vaccine is typical of a T-cell independent antigen, with an age-dependent
60 µg of each FHbp variant, 0.018 mg of polysorbate 80, and 0.25 mg response and poor immunogenicity in children younger than 2 years.
of Al3+ as AlPO4 in 10 mM histidine buffered saline at pH 6.0. The vaccine Little boost in antibody titer occurs with repeated doses; the antibody
is administered as a 0.5-mL dose intramuscularly. MenB-FHbp is licensed that is produced is relatively low-affinity IgM, and switching from IgM
as for a three-dose series and a two-dose series. In April 2016, the FDA to IgG production is poor.
approved a label change giving MenB-FHbp a flexible three-dose schedule Protective antibody concentrations are achieved within 10 to 14
of 0, 1 to 2 months, and 6 months and a two-dose schedule of 0 and 6 days of administration of MPSV4. The vaccine elicits protective levels
months. of bactericidal antibody to all four serogroups in more than 97% of
MenB-4C is a multicomponent vaccine consisting of three recom- recipients as measured at 28 days after vaccination. The antibody
binant proteins (i.e., neisserial adhesion A [NadA], factor H binding responses to each of the four polysaccharides in the polysaccharide
protein [FHbp] fusion protein, and neisserial heparin binding vaccine are serogroup specific and independent. Group A polysaccharide
antigen [NHBA] fusion protein) and outer membrane vesicles (OMVs) induces antibody in some children as young as 3 months, although a
containing outer membrane protein PorA serosubtype P1.4 (i.e., response comparable to that in adults is not achieved until the child is
New Zealand epidemic strain N298/254). The three recombinant 4 to 5 years of age.531 The serum antibody response to serogroup C is
proteins are individually produced in E. coli. The OMV antigenic age dependent, with a poor response in children younger than 2 years.303
component is produced by fermentation of N. meningitidis strain Serum concentrations of antibodies against group A and C polysac-
NZ98/254 (expressing outer membrane protein PorA serosubtype P1.4), charides decrease markedly during the first 3 years after receipt of a
followed by inactivation of the bacteria by deoxycholate, which also single dose of vaccine. The decrease in antibody occurs more rapidly
mediates vesicle formation. The antigens are adsorbed onto aluminum in infants and young children than in adults.384,712
hydroxide. Each 0.5-mL dose of MenB-4C contains 50 µg each of Field trials of A and C meningococcal vaccines in Europe and Africa
recombinant proteins NadA, NHBA, and FHbp; 25 µg of OMV; 1.5 mg have demonstrated efficacy rates against serogroup A of 85% to 95%
of aluminum hydroxide (0.519 mg of Al3+);, 3.125 mg of sodium 1 year after vaccination.531,667 After 3 years, efficacy rates were 67% for
chloride; 0.776 mg of histidine; and 10 mg of sucrose at pH 6.4 to older children but only 10% for children younger than 4 years at the
6.7. The vaccine is administered as a 0.5-mL dose intramuscularly. time of immunization with serogroup A vaccine.553 In an epidemic,
CHAPTER 245  Active Immunizing Agents 2569

serogroup C vaccine demonstrated clinical efficacy rates similar to those children achieved hSBA antibody titers of 1 : 8 or higher for meningococ-
for the serogroup A vaccine.633 cal serogroups C and Y, respectively, after the second dose.507
Serogroup Y and W135 antigens are immunogenic and safe in children When the MenACWY-D vaccine was licensed in 2005, some experts
older than 2 years. However, clinical efficacy has not been demonstrated predicted that the vaccine would be effective for up to 10 years, providing
for these preparations.12,57,664 protection through the period of highest risk in late adolescence and
People with deficiencies of the terminal components of serum early adulthood. Since the 2005 ACIP recommendations, additional
complement and those with anatomic or functional asplenia have data have improved our understanding of meningococcal conjugate
antibody responses to quadrivalent meningococcal vaccines consistent vaccines, including the duration of vaccine-induced immunity. Antibody
with protection.565,567 However, the clinical efficacy of vaccination has persistence studies indicate that circulating antibody declines 3 to 5
not been evaluated in these people. years after a single dose of MenACWY-D or MenACWY-CRM.296,388,665
Meningococcal conjugate vaccines prime the immune system, and Results from a vaccine effectiveness study demonstrate waning effective-
immunologic memory persists even in the absence of detectible bac- ness, and many adolescents are not protected 5 years after vaccination.203
tericidal antibodies. Although vaccine-induced immunologic memory ACIP concluded that a single dose of meningococcal conjugate vaccine
often protects against infection with other encapsulated bacteria such administered at age 11 or 12 years is unlikely to protect most adolescents
as Hib, detectable circulating antibody appears to be important for through the period of increased risk at ages 16 through 21 years. On
protection against N. meningitidis.304 Effectiveness of the three menin- the basis of this information, in 2011, ACIP recommended adding a
gococcal conjugate vaccines, which were licensed after MPSV4, was booster dose at 16 years of age.169
inferred by comparing SBA responses induced by the new vaccines MenB vaccine.  The immunogenicity of both serogroup B meningococ-
compared with SBA responses induced by MPSV4 in people 2 through cal vaccines (i.e., MenB-4C and MenBFHbp) has been evaluated in
55 years of age or by achieving a seroresponse at or above a predefined numerous published clinical trials.391,461,546,556,576,575,591,640 Both vaccines
bactericidal antibody titer among children 2 through 23 months of age. appear to provide short-term immunogenicity in healthy populations.
Protective antibody concentrations are achieved within 8 days after Studies on vaccine efficacy are not available. Licensure was based on
administration of MenACWY-D.78 In studies conducted among people the ability of the vaccines to elicit detection of bactericidal antibody
11 to 55 years of age comparing the immunogenicity of conjugate vaccine that is presumed to indicate protection. Studies regarding antibody
with that of the polysaccharide vaccine at 28 days after vaccination, the persistence are limited. Immunogenicity studies in populations at
percentage of those achieving at least a fourfold increase in bactericidal increased risk for invasive meningococcal disease have not been
titer for each serogroup was higher in the MPSV4 group than in the completed.
MenACWY-D group for people older than 18 years. Nonetheless, the MenB-FHbp has been administered concomitantly with the following:
criteria for demonstrating immunologic noninferiority to MPSV4 were quadrivalent human papillomavirus (HPV) vaccine591 but not 9-valent
still achieved. The percentage of those with at least a fourfold rise was HPV vaccine, with MenACWY (unpublished data), and with Tdap
highest for serogroup W135 and lowest for serogroup Y. The percentage vaccine (Adacel, Sanofi Pasteur [unpublished data]).
of people achieving a protective level of bactericidal antibody was greater
than 97% for all serogroups in the MenACWY-D and MPSV4 groups. Adverse Events
Response to revaccination with MenACWY-D was assessed by MenACWY vaccines.  MPSV4 has been used extensively in mass-
administering MenACWY-D to people previously vaccinated with MPSV4 immunization programs and in the military and among international
or MenACWY-D and to vaccine- naïve control subjects. All people in all travelers. Adverse reactions to MPSV4 usually are mild; the most frequent
three groups achieved protective bactericidal antibody titers at 8 and 28 reactions are pain and redness at the injection site that last for 1 or 2
days after receiving MenACWY-D. Those initially primed with MenACWY- days.62 Estimates of the incidence of local reactions range from 4% to
D achieved higher bactericidal antibody concentrations than those of 56%. Transient fever occurs in as many as 5% of vaccine recipients in
the naïve controls for all serogroups except A. In contrast, bactericidal some studies but is less common in older children and adults.
antibody titers of those primed with MPSV4 were lower than those of The most frequently adverse events reported to VAERS for MenACWY-
vaccine-naïve controls on days 8 and 28 for all serogroups. D include fever (16.8%), headache (16.0%) injection-site erythema
In study participants 11 to 18 years of age, noninferiority of (14.6%), and dizziness (13.4%). Syncope has been identified as an adverse
MenACWY-CRM to MenACWY-D was demonstrated for all four event after any vaccination, with a higher proportion of syncope events
serogroups.166 The proportions of people with seroresponses were statisti- reported to VAERS occurring in adolescents than other age groups.154
cally higher for serogroups A, W, and Y in the MenACWY-CRM group, Syncope was listed in 10% of reports involving MenACWY-D. Twenty-
compared with the MenACWY-D group. The clinical relevance of higher four deaths (0.3%) were reported.
postvaccination immune responses is not known. The most frequently reported adverse events for MenACWY-CRM
The immunogenicity of MenACWY-CRM in children 2 through 10 were injection-site erythema (19.7%) and injection site swelling (13.7%).
years of age was evaluated in a multicenter, randomized, controlled Syncope was listed in 8.8% of reports involving MenACWY-CRM. One
trial.327 After a single MenACWY-CRM dose, seroresponses to group death (0.4%) was reported.
C, Y, and W135 in children 2 through 5 years and 6 through 10 years Rates of local and systemic adverse events observed after administra-
of age were noninferior to responses after a single MenACWY-D dose. tion of Hib-MenCY-TT were comparable to rates observed after
MPSV4 is administered subcutaneously, whereas MCV4 is admin- administration of Hib-TT. Hib-MenCY-TT was found to be safe and
istered intramuscularly. More than 100 people have inadvertently received immunogenic for Hib and meningococcal serogroups C and Y.
the MCV4 vaccine by the subcutaneous route. For a subset of these MenB vaccine.  The safety of both serogroup B meningococcal vaccines
individuals, the CDC determined that although the serologic responses (i.e., MenB-4C and MenBFHbp) have been evaluated in numerous
were lower after MCV4 was administered subcutaneously compared published clinical trials.391,461,556,575,591,597 There were no deaths considered
with intramuscularly, the proportions of individuals who achieved to be related to either vaccine. The most frequently reported severe
antibody levels thought to be protective were similar. The CDC therefore local and systemic adverse events for MenB-4C include injection site
did not recommend that those who had received MCV4 needed to be pain (20–29%), fever of 38°C or higher (1–5%), headache (4–6%),
reimmunized.149 fatigue (4–6%), myalgias (12–13%), and joint pain (2%). For MenBFHbp,
In clinical trials Hib-MenCY-TT was coadministered with DTaP- the most frequently reported severe local and systemic adverse events
HepB-IPV and 7-valent pneumococcal conjugate vaccine (PCV7) at include injection site pain (5–8%), fever of 38°C (2–8%), headache
ages 2, 4, and 6 months and with MMR, varicella, and PCV7 vaccines (1%), fatigue (1–4%), myalgias (1–3%), and joint pain (1%).
at 12 to 15 months of age. No decreased immunogenicity of coadmin-
istered vaccines was observed.96,460 A randomized, controlled, multicenter Indications
study evaluated the percentage of subjects with hSBA titers of 1 : 8 or Meningococcal polysaccharide vaccine.  Routine vaccination of civilians
higher at 2 months after the second dose was administered at 4 months with MPSV4 is not recommended.204 Use of MPSV4 should be limited
of age. In the group vaccinated with Hib-MenCY-TT, 94% and 83% of to people older than 55 years or when MenACWY is not available.
2570 PART V  Prevention of Infectious Diseases

TABLE 245.13  Recommended Meningococcal TABLE 245.14  Meningococcal Vaccine


Vaccines for Immunocompetent Children Recommendations by Risk Group
Age Vaccine Status MenACWY Vaccines MenB Vaccines
2 mo–10 y MenACWY-Da Not routinely recommended Complement deficiencya Complement deficiencya
MenACWY-CRMb Anatomic or functional aspleniab Anatomic/functional aspleniab
HibMenCY-TTc Outbreakc Outbreakc
10–25 y MenB-FHbp Not routinely recommended Microbiologistsd Microbiologistsd
or HIV infection
MenB-4C Travelerse
11–21 y MenACWY-D Primary: First-year college studentsf
or Age 11–12 y: 1 dose
Military recruits
MenACWY-CRM Age 13–18 y: 1 dose if not
previously immunized a
Inherited or chronic deficiencies of C3, C5–C9, properdin, factor D, or factor H or those
Age 19–21 y: not routinely receiving eculizumab.
b
recommended but may be given as Includes sickle cell disease.
c
catch-up immunization for those The CDC defines outbreaks and those at risk.
d
who have not received a dose Only microbiologists who routinely work with N meningitidis.
e
To areas with hyperendemic or epidemic meningococcal disease.
after their 16th birthday f
Unvaccinated or inadequately vaccinated first-year college students who live in
Booster: residence halls.
1 dose recommended for adolescents Modified from Committee on Infectious Diseases, American Academy of Pediatrics.
if first dose administered before Recommendations for serogroup B meningococcal vaccine for persons 10 years and older.
16th birthday Pediatrics. 2016;138:e20161890.
a
Licensed only for people 9 mo to 55 y.
b
Licensed only for people 2 mo to 55 y.
c
Licensed only for children 6 wk to 18 mo. children with meningococcal conjugate vaccines are shown in Table
Modified from American Academy of Pediatrics. Meningococcal infections. In: Kimberlin 245.15.28,39,204,445
DW, Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on MenB vaccines.  Routine vaccination with meningococcal serogroup
Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; B vaccine is recommended for those 10 years or older who are at increased
2015:547–58. risk for serogroup B disease (see Tables 245.14 and 245.15), including
the following groups: (1) people with persistent complement component
diseases, including inherited or chronic deficiencies in C3, C5-C9,
Meningococcal conjugate vaccines.  Routine childhood immunization properdin, factor D, or factor H, and people receiving eculizumab; (2)
with meningococcal conjugate vaccine (i.e., MenACWY) is recommended people with anatomic or functional asplenia, including sickle cell disease;
for adolescents 11 through 18 years of age, but it is not recommended and (3) healthy people who are at increased risk because of a serogroup
for children 2 months through 10 years of age because the infection B meningococcal disease outbreak.211,276,444 The AAP and the CDC do
rate is low for this age group; the immune response is less robust than not express a preference for the two licensed vaccines, but the same
in older children, adolescents, and adults; and duration of immunity product must be used for the entire series.
is unknown.28,39,204 Recommendations for routine use of MenACWY in A three-dose schedule for MenB-FHbp is recommended for people
adolescents are shown in Table 245.13 and are as follows:28,39,204 at increased risk for meningococcal serogroup B disease. For healthy
• Adolescents should be immunized routinely at the 11- to 12-year people 16 to 23 years of age who are not at increased risk for meningococ-
of age health care visit, when immunization status and other preven- cal serogroup B disease, the ACIP has a permissive recommendation
tive health services can be addressed. A booster dose at 16 years of that allows use of one of the meningococcal serogroup B vaccines if
age is recommended for adolescents immunized at 11 through 12 the patient desires to be immunized.211,444 If an adolescent so chooses
years of age. and MenB-FHbp is selected, the ACIP recommends a two-dose schedule.
• Adolescents 13 through 18 years of age should be immunized routinely If a patient receives a second dose of MenB-FHbp less than 6 months
with a meningococcal conjugate vaccine if not previously immunized. after the first dose, a third dose should be given at least 6 months after
Adolescents who receive the first dose at 13 through 15 years of age the first.
should receive a one-time booster dose at 16 through 18 years of
age. Postexposure Immunoprophylaxis
• Adolescents who receive their first dose of meningococcal conjugate Because secondary cases can occur several weeks or more after onset
vaccine at or after 16 years of age do not need a booster dose unless of disease in the index case, meningococcal vaccine is an adjunct to
they have risk factors. chemoprophylaxis when an outbreak is caused by a serogroup prevented
• People 19 to 21 years of age are not routinely recommended to by a meningococcal vaccine. For control of meningococcal outbreaks
receive meningococcal conjugate vaccine, but vaccine may be caused by vaccine-preventable serogroups (i.e., A, C, Y, and W135), the
administered as a catch-up dose for those who have not received a preferred vaccine for adults and children 2 years and older is a menin-
dose after their 16th birthday. gococcal conjugate vaccine (i.e., MenACWY).204 For outbreaks caused
The ACIP and the AAP recommend routine use of meningococcal by serogroup B meningococcal disease, vaccination with a serogroup
conjugate vaccines (e.g., MenACWY) for people 2 months or older who B meningococcal vaccine is recommended for those 10 years or older
have certain medical conditions that increase the risk of meningococcal identified as being at increased risk because of the outbreak.178
disease, including those who have persistent (e.g., genetic) deficiencies
in the complement pathway (e.g., C3, properdin, factor D, factor H, Precautions and Contraindications
C5-C9), people receiving eculizumab (because the drug binds C5 Vaccination with MenACWY, MPSV4, or Hib-MenCY-TT is contrain-
and inhibits the terminal complement pathway), and people with dicated for people known to have a severe allergic reaction to any
functional or anatomic asplenia, including people with sickle cell component of the vaccine, including diphtheria or tetanus toxoid. The
disease (Table 245.14).28,39,204 In 2016, the ACIP recommended ACIP does not consider a history of Guillain-Barré syndrome to be a
that people 2 months or older with HIV infection also be given menin- contraindication or precaution for meningococcal vaccination.204 Because
gococcal conjugate vaccines based on data indicating an increased risk MenACWY, MPSV4, and Hib-MenCY-TT are inactivated vaccines, they
of disease.445 Specific recommendations for immunization of high-risk can be administered to people who are immunosuppressed as a result
CHAPTER 245  Active Immunizing Agents 2571

TABLE 245.15  Recommended Immunization Schedule and Intervals for Children at Risk for
Invasive Meningococcal Diseasea
Age Subgroup Primary Immunization Booster Doseb
c
2–18 mo Children who have persistent complement 4 doses of HibMenCY-TT at 2, Person remains at increased risk and
deficiencies, have functional or anatomic asplenia 4, 6, and 12–15 moc first dose received at following age:
are at risk during a community outbreak For 2 mo–6 y of age: should receive
attributable to a vaccine serogroup additional dose of MenACWY 3 y after
2–23 mo with Children who have persistent complement 4 doses of MenACWY-CRM primary immunization; boosters should
high-risk conditions deficiencies, have functional or anatomic asplenia, (Menveo) at 2, 4, 6, and be repeated every 5 y thereafter
have HIV, travel to or are residents of countries 12 mo For ≥7 y of age: should receive
where meningococcal disease is hyperendemic or additional dose of MenACWY 5 y after
epidemic are at risk during a community outbreak primary immunization; boosters should
attributable to a vaccine serogroup be repeated every 5 y thereafter
9–23 mo with Children who have persistent complement 2 doses of MenACWY
high-risk conditions deficiencies, travel to or are residents of countries (Menactra), 12 wk apartd
where meningococcal disease is hyperendemic or
epidemic are at risk during a community outbreak
attributable to a vaccine serogroup
2–55 y with high-risk People who have persistent complement deficiencies, 2 doses of MenACWY, 8–12 wk
conditions and not have functional or anatomic asplenia, have HIV apart
immunized People who travel to or are residents of countries 1 dose of MenACWYe
previouslye,f where meningococcal disease is hyperendemic or
epidemic are at risk during a community outbreak
attributable to a vaccine serogroup
≥10 y with high-risk People who have persistent complement deficiencies, 2 doses of MenB-4C 4 wk apart —
conditions have functional or anatomic asplenia are at risk or
during a community outbreak attributable to a 3 doses of MenB-FHbp
vaccine serogroup administered at 0, 2 and 6 mo
a
Includes children who have persistent complement deficiencies (e.g., C5–C9, properdin, factor H or factor D) and anatomic or functional asplenia; travelers to or residents of countries in
which meningococcal disease is hyperendemic or epidemic; and children who are part of a community outbreak of a vaccine-preventable serogroup.
b
If child remains at increased risk of meningococcal disease.
c
Infants and children who received Hib-MenCY-TT and are traveling to areas with highly endemic rates of meningococcal disease, such as the African “meningitis belt,” are not protected
against serogroups A and W and should receive a quadrivalent meningococcal vaccine licensed for children age ≥2 mo before travel.
d
Because of high risk of invasive pneumococcal disease, children with functional or anatomic asplenia should not be immunized with MenACWY-D (Menactra) before age 2 y to avoid
interference with the immune response to the pneumococcal conjugate vaccine (PCV) series.
e
If MenACWY-D is used, administer at least 4 weeks after completion of all PCV doses.
f
If an infant is receiving the vaccine before travel, two doses can be administered as early as 2 months apart.
Modified from American Academy of Pediatrics. Meningococcal Infections. In: Kimberlin DW, Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious
Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:547–58.

of disease or medications; however, response to the vaccine may be less cases.111,199 The resurgence was attributed to a susceptible cohort of
than optimal. adolescents and young adults entering high school and college who
No RCTs have been conducted primarily to evaluate use of MPSV4 were not targeted for vaccination and spared from natural infection by
or MenACWY vaccines in pregnant or lactating women. VAERS reports declining disease rates. In 1989, the ACIP and AAP implemented a
of exposure to MPSV4 during pregnancy have not identified adverse two-dose combined MMR schedule given at 4 to 6 or at 11 to 12 years
effects among pregnant women or newborns of women vaccinated of age.110 By the early 2000s, on average, fewer than 270 cases were
during pregnancy. From the VAERS reports available for women found reported annually, and seasonal peaks were no longer seen.67
to be pregnant at the time of MenACWY-D vaccination, no major Before vaccine licensure in 1967 and during the early years of vaccine
safety concerns associated with vaccination have been identified in the use, most reported cases occurred in the 5- to 9-year age group, with
mother or fetus. Pregnancy should not preclude vaccination with 90% of cases occurring among children 15 years of age and younger.
MenACWY or MPSV4, if indicated.204 Women of childbearing age who In the late 1980s, a shift toward older children occurred. Since 1990,
become aware that they were pregnant at the time of MenACWY vac- people 15 years or older have accounted for 30% to 40% of cases per
cination should contact their health care provider or the vaccine year.
manufacturer so that their experience can be captured in the manu- In 2006, an outbreak of 6584 cases occurred and was centered on
facturer’s registry of vaccination during pregnancy. highly two-dose vaccinated college students in the American Midwest.229
Before administering MenB vaccines, treating clinicians should consult The 2006 outbreak underscored limitations in the 1998 recommendations
the package insert for a full list of precautions, warnings, and contra- about prevention of mumps transmission in health care and other
indications.211,444 Pregnancy and breastfeeding are precautions because settings with a high risk of mumps transmission. After reviewing data
neither vaccine has been evaluated in these situations. A severe allergic from this outbreak and previous evidence of mumps vaccine effectiveness
reaction to a previous dose of MenB vaccine or any of its components and transmission, the ACIP issued updated recommendations, specifying
is a contradiction. that all children and adults in certain high-risk groups, including students
at post–high school educational institutions, health care personnel, and
Mumps Vaccine international travelers, should receive two doses of mumps-containing
Live virus mumps vaccine became available in the United States in 1967 vaccine.110 In the next 2 years, the number of reported cases returned
and was recommended for routine use in 1977. The incidence of reported to usual levels, and outbreaks involved fewer than 20 cases.
mumps cases decreased rapidly after vaccine licensure until 1986 through Outbreaks have continued to occur, particularly in high-density,
1987, when a resurgence of mumps occurred resulting in 20,638 reported close-contact settings, despite high two-dose coverage. Because two
2572 PART V  Prevention of Infectious Diseases

doses of mumps-containing vaccine are not 100% effective, most mumps among populations with high two-dose coverage found two doses of
cases in settings of high immunization coverage such as the United mumps-containing vaccine to be 80% to 92% effective in preventing
States most often occur among people who have received two doses. clinical disease.452,581 In the 1988 through 1989 outbreak among junior
In the 2009–10 season, a large outbreak occurred in the northeastern high school students, the risk of mumps was five times higher for students
United States, affecting more than 3500 people, primarily members of who received one dose compared with students who received two doses.341
traditional observant communities in New York and New Jersey.163 There Population and school-based studies in Europe and Canada estimate
was also an outbreak in the US territory of Guam, with 505 cases two doses of mumps-containing vaccine to be 66% to 95% effec-
reported.500 Most patients had received two doses of MMR vaccine and tive.104,202,233,241,335,578,612 Despite relatively high two-dose vaccine effective-
were exposed in communal settings. In 2011 through 2013, there were ness, high two-dose vaccine coverage may not be sufficient to prevent
several smaller mumps outbreaks reported on college campuses in all outbreaks.5,216,229
California,173 Virginia, and Maryland. However, they had limited spread, Studies indicate that one dose of MMR vaccine can provide persistent
and national case counts for these years were at several hundred cases antibodies to mumps. Most people (70–99%) examined approximately
per year. 10 years after initial vaccination had detectable mumps antibodies.92,306,422
In 2014, several outbreaks affiliated with universities were reported T-lymphocyte immunity to mumps was found in 70% of adults vac-
from multiple states, including one community outbreak in Ohio linked cinated in childhood compared with 80% of adults who acquired a
to a university that involved over 400 people and an outbreak affecting natural infection during childhood.213,332 In two-dose recipients, mumps
the National Hockey League. Outbreaks have been reported from several antibodies were detectable in 74% to 95% of people followed over 12
university campuses in 2015 through 2016, including a number of smaller years after receipt of a second dose of MMR vaccine, but antibody levels
outbreaks with limited spread. The two largest outbreaks were from declined with time.227,422 Mumps antigen–specific lymphoproliferative
Iowa and Illinois, each involving several hundred university students; responses have been detected in vaccine recipients without detectable
both held wide-scale vaccination campaigns that included providing a mumps antibodies, but the role of these responses in protection against
third dose of MMR as a control measure.7 mumps disease is not clear.377,653
In 2016, the greatest number of mumps cases since the outbreaks
of 2006 was reported. These outbreaks involved 45 states and more Adverse Events
than 2800 individuals. Six states (i.e., Arkansas, Illinois, Indiana, Iowa, The use of mumps vaccine is associated with very few side effects.
Massachusetts, and Oklahoma) reported more than 100 cases in 2016. Parotitis and fever have been reported rarely. Hypersensitivity reactions,
including rash, pruritus, and purpura, have been associated temporally
Preparations with vaccination, but they are transient and usually mild. Administration
The live attenuated mumps virus vaccine (i.e., Jeryl Lynn strain) used of MMR is not harmful if it is given to an individual already immune
in the United States is prepared in chick embryo cell culture and is to one or more of the viruses.40,471
available in combination as MMR and MMRV. The AAP and ACIP The frequency of reported central nervous system dysfunction after
recommend that combined MMR vaccine be used when any of the vaccination is not greater than the observed background rate for unim-
individual components is indicated, and MMRV is recommended if munized people.471 The IOM concluded that evidence is inadequate to
the vaccinee is 12 months through 12 years of age. establish a causal relationship between the Jeryl Lynn strain of mumps
Each dose of vaccine contains neomycin, sorbitol, and hydrolyzed vaccine used in the United States and aseptic meningitis, encephalitis,
gelatin as stabilizers. Before reconstitution, MMR vaccine must be stored or sensorineural deafness.362
at 2°C to 8°C (35.6°F–46.4°F) or colder and protected from light to
avoid inactivation. After reconstitution, the vaccine should be used Indications
within 8 hours or discarded. Routine active immunization as MMR of children 12 to 15 months of
age is recommended.40,471 Most children receive a second dose of mumps
Immunogenicity and Efficacy vaccine in childhood as a result of the recommendation for routine
The vaccine induces an asymptomatic, noncommunicable infection. measles revaccination with MMR. Susceptible older children, adolescents,
After vaccination with MMR vaccine, approximately 94% of infants and adults also should be vaccinated against mumps.
and children develop detectable mumps antibodies with (range, Evidence of immunity through documentation of vaccination is
89–97%).85,254,264,396,425,544,548,588,589,647,648 Vaccination induces relatively low defined as one dose of live mumps vaccine for preschool-aged children
levels of antibodies compared with natural infection.347,675 After a second and adults not at high risk for exposure and infection and two doses
dose of MMR vaccine, most people mounted a secondary immune of live mumps vaccine for school-aged children (i.e., kindergarten to
response, approximately 50% had a fourfold increase in antibody titers, grade 12) and adults at high risk for exposure and development of
and the proportion with low or undetectable titers was significantly infection (i.e., health care workers, international travelers, and students
reduced from 20% before vaccination with a second dose to 4% at 6 at post–high school education institutions). Additional recommendations
months after vaccination.92,306,422 for outbreak control include administering a second dose of MMR for
Although antibody measurements are often used as a surrogate measure preschool-aged children and adults not at high risk for exposure and
of immunity, no serologic tests are available for mumps that consistently acquisition of infection if these people are part of a group that is
and reliably predict immunity. The immune response to mumps vaccina- experiencing an outbreak.150
tion probably involves the humoral and cellular immune response, but To ensure high levels of immunity, especially among groups at high
no definitive correlates of protection have been identified. risk for exposure and development of infection, every opportunity
Clinical studies conducted before vaccine licensure involving should be used to provide the first or second dose of MMR vaccine to
approximately 7000 children found a single dose of mumps vaccine to those without adequate evidence of immunity (e.g., documentation of
be approximately 95% effective in preventing mumps disease.348,624,675 vaccination). No formal recommendation for a third MMR dose exists,
However, vaccine effectiveness estimates have been lower in postlicensure but the CDC has provided guidelines for public health agencies consider-
studies. The median vaccine effectiveness against mumps has been ing its use as a control measure during mumps outbreaks.266 Factors
estimated at 78% for one dose and 88% for two doses.471 In the United that can trigger a recommendation include outbreaks in populations
States, estimated vaccine effectiveness for one dose of mumps-containing with two-dose MMR vaccination coverage of greater than 90%, intense
vaccine is between 81% and 91% in junior high and high school set- exposure settings such as universities, evidence of sustained transmission
tings180,183,341,389,626,684 and between 64% and 76% among household or for greater than 2 weeks, and high attack rates (>5 cases/1000 people).
close contacts.452,626 Population and school-based studies conducted in Mumps remains endemic throughout most of the world. Although
Europe and Canada report comparable estimates of 49% to 92% for vaccination against mumps is not a requirement for entry into any
vaccine effectiveness.104,181,202,233,241,335,431,555,584,612,641 country, susceptible children, adolescents, and adults born after 1956
Fewer studies have been conducted to assess the effectiveness of two should be offered mumps vaccination, usually as MMR, before engaging
doses of mumps-containing vaccine. In the United States, outbreaks in international travel.
CHAPTER 245  Active Immunizing Agents 2573

Mumps vaccine has no proven value in the prevention of Data from the National Notifiable Disease Surveillance System reveal
disease in susceptible individuals after exposure to mumps, probably an escalation in the increase in all age groups between 2001 and 2004.90
because the time required to develop protective antibody titers after Incidence rates for the pediatric population peak for infants younger
immunization exceeds the incubation period of clinical mumps. However, than 6 months and again during adolescence between 11 and 18 years
if the exposure does not result in infection, the vaccine confers subsequent of age.21 The increase in the number of cases among young infants
immunity. suggests that a true increase in pertussis circulation has occurred. The
number of cases among children old enough to receive vaccine has
Precautions and Contraindications remained stable. The likely reason that pertussis remains endemic in
For people who are allergic to eggs, the risk of serious allergic reactions the United States despite high vaccination rates is that neither infection
such as anaphylaxis after receiving MMR is extremely low, and skin nor immunization provides life-long immunity.
testing with vaccine cannot predict an allergic reaction to vaccina- Humans are the only known reservoir for pertussis. Vaccine effective-
tion.40,369,386 Performing skin testing is not required before administering ness falls off rapidly after immunization, with rates of 68% after the
MMR to people who are allergic to eggs. Similarly, the administration first year dropping to 8.9% after 4 years.393 Pregnant woman who have
of gradually increasing doses of vaccine is not required.471 Data indicate received Tdap in the third trimester of pregnancy had significant declines
that most anaphylactic reactions to measles- and mumps-containing in immunity 9 to 15 months after delivery.1
vaccines are not associated with hypersensitivity to egg antigens but The experiences of countries where rates of pertussis vaccination
rather with hypersensitivity to other components of the vaccines, such have markedly declined provide strong support for continuing routine
as the gelatin stabilizer.311,340,385,419 immunization of infants and young children.188 In the United Kingdom,
Because of the theoretical risk of fetal damage, mumps vaccine should as a result of adverse publicity about pertussis vaccination, a decrease
not be administered to women known to be pregnant or who are in immunization rates for 2-year-old children from 77% in 1974 to
considering becoming pregnant. Women vaccinated with MMR should 30% in 1978 was followed by an epidemic of 102,500 cases of pertussis.
avoid conception for 28 days after vaccination.471 A similar experience occurred in Japan, which reported 13,105 cases
Lymphoreticular or other generalized malignancy and primary or and 41 deaths in 1979 after routine immunization had been suspended
secondary immunodeficiency states represent contraindications to the temporarily in 1975.
use of mumps vaccine. Exceptions are children with HIV infection who In the United States during the 1980s, publicity about alleged serious
are immunized against measles with MMR (see Measles Vaccine). Because reactions to pertussis vaccine generated public controversy about the
infection after vaccination is noncommunicable, susceptible close contacts risk of receiving pertussis vaccine. This resulted in costly litigation and
of immunosuppressed patients should be vaccinated to avoid exposure escalating vaccine costs and jeopardized vaccine supply and develop-
to mumps in these patients. ment.490,532 The experience in countries such as England and Japan, the
After cessation of immunosuppressive therapy, mumps vaccine usually severity of pertussis in young infants, and the usually benign or self-
is withheld for at least 3 months. Because the intensity and type of limited sequelae of pertussis vaccination clearly justify continuing routine
immunosuppressive therapy, radiation therapy, underlying disease, and childhood immunization. Several risk-benefit analyses provided
other factors determine when immunologic responsiveness will be additional evidence in support of vaccination.188,349
restored, making a definitive recommendation for an interval after Effective primary preventive programs necessitate immunizing young
cessation of immunosuppressive therapy when mumps vaccine can be infants, usually beginning at 2 months of age, because the morbidity
safely and effectively administered often is not possible. and mortality of pertussis are greatest for infants, especially those younger
The effect of immune globulin preparations on the response to than 6 months.123,188 Approximately 26% of reported cases in the United
mumps vaccine is unknown. High doses of immune globulin preparations States occur in infants younger than 6 months. During the period from
can inhibit the immune response to measles vaccine for 3 or more 1997 through 2000, the case-fatality rate for infants younger than 6
months, depending on the dosage.29 If mumps vaccine is given as the months was 0.8%; 63% of these infants were hospitalized with frequent
MMR vaccine, the recommendations for measles vaccine should be complications, including pneumonia (11.8%), seizures (1.4%), and
followed (see Table 245.11). encephalopathy (0.2%).142 From 2000 through 2004, 100 pertussis-related
Administration of mumps vaccine should be avoided if the individual deaths were reported; 90% were among infants younger than 4 months,
is receiving immunosuppressive dosages of systemic corticosteroids. and 76% occurred in infants younger than 2 months.90
The effects of corticosteroids vary, but many clinicians consider that a In 2014, 23,971 cases of pertussis were reported to the CDC; 9935
dose equivalent to 2 mg/kg per day or 20 mg/day of prednisone is cases occurred during an outbreak in California to produce a rate of 26
sufficiently immunosuppressive to raise concern about the safety of cases per 100,000 people, with a rate of 1746 cases per 100,000 infants
vaccination with live virus vaccines. younger than 12 months. Three deaths were attributed to this outbreak,
all were infants with an onset of illness at less than 5 weeks of age.691
Pertussis Vaccine Maintaining high rates of immunization in children beyond infancy,
Pertussis (i.e., whooping cough), which is caused by the fastidious, with booster immunization for adolescents and adults, may reduce the
gram-negative, pleomorphic bacillus B. pertussis, continues to produce risk of infection in infants by decreasing the incidence of infection in
significant morbidity and mortality worldwide among young children.491 older family members and the resultant transmission of B. pertussis
In the absence of vaccination, the WHO estimated that approximately within the household. Vaccination, including boosters, is essential because
1 million deaths would have occurred from the disease and its complica- the disease is highly infectious, and transmission often occurs before
tions. In the United States, the number of cases has been reduced by adults seek medical care.
approximately 95% during the vaccine era.
Despite an effective vaccine, pertussis continues to occur in the Preparations
United States in all age groups. Since the historic low point achieved Whole-cell pertussis vaccine was introduced in the United States in the
in the 1980s, the incidence of pertussis has increased steadily. Outbreaks 1920s. Not until the 1940s to 1950s, however, did pediatric whole-cell
have occurred recently in California, Washington, and Minnesota. In pertussis vaccine become routinely recommended for children. This
the California outbreak, there was a degree of clustering within popula- vaccine, unavailable in the United States since 2002, consisted of a
tions for whom vaccination rates, largely due to nonmedical exemptions, suspension of inactivated B. pertussis combined with diphtheria and
were low.60 Nonmedical exemption laws appear to increase the rates of tetanus toxoids (DTP).90
pertussis in local areas of undervaccination in the unvaccinated and To reduce the incidence of local and systemic reactions caused by
vaccinated population.60,706 whole-cell vaccines, less reactogenic pediatric acellular vaccines composed
Surveillance data collected by the CDC’s National Immunization of one or more purified components of B. pertussis combined with
Program for the periods from 1994 through 1996 and 1997 through diphtheria and tetanus toxoids (DTaP) were developed. Initially licensed
2000 demonstrated that the incidence of pertussis increased 60% among in 1991 for use as the fourth or fifth dose in the series, DTaP was licensed
adolescents and adults and 11% among infants younger than 6 months.142 in 1997 for all five doses in the series.
2574 PART V  Prevention of Infectious Diseases

TABLE 245.16  Acellular Pertussis Vaccines Licensed for Use in the United States
Trade Vaccine No. of Pertussis Antigenic
Vaccine Name Manufacturer Antigens Content Dose Series Approved
DTaP Vaccines for Children Age <7 Years
DTaP Daptacel Sanofi Pasteur 5 PT, FHA, PE, 2 All 5 doses
types of FIM
DTaP Infanrix GlaxoSmithKline Biologicals 3 PT, FHA, PE All 5 doses
DTaP-IPV Kinrix GlaxoSmithKline Biologicals 3 PT, FHA, PE Booster dose for 5th dose of DTaP and 4th dose
of IPV, children 4 through 6 years of age
DTaP-hepatitis B-IPV Pediarix GlaxoSmithKline Biologicals 3 PT, FHA, PE First 3 doses, children 6 wk–6 y of age
DTaP-IPV/Hib Pentacel Sanofi Pasteur 5 PT, FHA, PE, 2 First 4 doses, children 6 wk–4 y of age
types of FIM
DTaP-IPV Quadracel Sanofi Pasteur 5 PT, FHA, PE, 2 Booster dose for 5th dose of DTaP and 4th dose
types of FIM of IPV, children 4–6 y of age
Tdap Vaccines for Adolescents
Tdap Adacel Sanofi Pasteur 5 PT, FHA, PE, 2 Single dose at 11–12 y of age instead of Td
types of FIM
Tdap Boostrix GlaxoSmithKline Biologicals 3 PT, FHA, PE Single dose at 11–12 y of age instead of Td
DTaP, Diphtheria–tetanus–acellular pertussis; FHA, filamentous hemagglutinin; FIM, fimbriae; Hib, Haemophilus influenzae type b vaccine; IPV, inactivated poliovirus vaccine; PE, pertactin;
PT, pertussis toxin; Td, tetanus and reduced diphtheria toxoids; Tdap, tetanus–diphtheria–acellular pertussis.
Modified from American Academy of Pediatrics. Pertussis (whooping cough). In: Kimberlin DW, Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious
Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:608–21.

Numerous acellular vaccines formulated from the different compo- epidemiologic studies must be performed to demonstrate efficacy. Studies
nents have been tested in children. All currently US licensed vaccines in the United States of household contacts exposed to pertussis indicate
contain detoxified or inactivated pertussis toxin (i.e., pertussis toxoid) that the efficacy of whole-cell vaccine was 80% or greater.123,188,513 Studies
and filamentous hemagglutinin.231 Most vaccines include one or both reporting lower rates of vaccine efficacy often reflect the use of different
of the following B. pertussis antigens: pertactin (PRN, a 69-kd outer criteria for the diagnosis of pertussis and lesser effectiveness of the
membrane protein) and fimbriae proteins (FIM, agglutinogens). Various vaccine in protecting against mild infection than against severe disease.268
vaccines have different amounts of each component. No pertussis-only Vaccine-induced immunity persists for at least 3 years and subsequently
vaccine is available. diminishes with time.
From 2001 until 2008, numerous changes were made to the list of Pertussis in individuals previously vaccinated is less severe and is
licensed acellular pertussis–containing vaccines available in the United associated with fewer complications than in unvaccinated people. In a
States. Several acellular vaccines are available for use in the primary case-control study of pertussis, immunization of both parents decreased
vaccination series for children younger than 7 years (Table 245.16). the incidence of disease by 51% among infants younger than 4 months.691
For enhanced control of pertussis, routine revaccination of adolescents Vaccine effectiveness in pregnant women is 91%.50
and adults with an acellular pertussis vaccine is recommended to In studies of the efficacy of eight acellular pertussis vaccines in
minimize the morbidity associated with infection in these age groups infants, rates of prevention of pertussis ranged from 58% to 93%.541
and to reduce the reservoir of infection. Tdap vaccines contain the Comparing efficacy of the different products, however, often is not
same pertussis antigen components as are in the pediatric vaccines possible because of differences in study design, vaccine schedule (i.e.,
(some in reduced quantities). The tetanus and diphtheria components number of doses and age of administration), case definitions of pertussis,
are the same as for the current adult formations of Td, with reduced and other confounding variables. The acellular vaccines appear to be
diphtheria content as recommended for use in people 7 years of age similar in efficacy to most whole-cell vaccines. Whereas in two large
or older. trials in Sweden and Italy, several acellular vaccines demonstrated
The ACIP recommends that whenever feasible, the same DTaP vaccine substantially greater efficacy than seen in the one US whole-cell vaccine
product should be used for all doses of the vaccination series. If the previously approved, other whole-cell vaccines studied appeared to be
vaccine provider does not know or does not have available the type of slightly more effective than acellular vaccines in other trials.252 The
DTaP previously administered, any of the available licensed DTaP vaccines vaccines in the Swedish and Italian trials were given in a three-dose
can be used to complete the vaccination series.138 schedule, in contrast to the four-dose primary schedule for vaccination
of young children in the United States.
Immunogenicity
The Multicenter Acellular Pertussis Trial evaluated the immunogenicity Adverse Events
and safety of 13 acellular pertussis–containing vaccines compared with Local and febrile reactions to whole-cell vaccines occur in more than
whole-cell DTP administered to infants at 2, 4, and 6 months of age one half of DTP recipients.200 These manifestations usually develop
in the United States.231 Serologic correlates of immunity to diphtheria within the first 24 hours and are brief in duration. More serious reactions
and tetanus toxoids, defined as antibody levels 0.1 IU/mL or greater, to whole-cell vaccines are uncommon. The reactions include prolonged
were achieved after vaccination with acellular vaccines and were crying for 3 hours or longer occurring within the first 48 hours of
comparable with those achieved with DTP vaccination. Pertussis receiving vaccination (1% to 3% of DTP recipients) and a temperature
immunity, determined by at least a fourfold rise in antibody titer to of 40.5°C or greater (≥104.9°F) within 48 hours (0.3%); a hypotonic-
pertussis toxin and filamentous hemagglutinin, was equal to or greater hyporesponsive episode described as collapse or shock-like state within
than results with DTP after immunization with either of the two DTaP 48 hours and seizure within 3 days of vaccination was estimated to
vaccines.253 occur once per 1750 doses.200 Episodes of inconsolable crying, high
fever, and hypotonic-hyporesponsive episodes after receipt of DTP
Efficacy vaccine resolved without sequelae.
For whole-cell and acellular vaccines, serologic correlates of immuno- Most post-DTP seizures occurring within 48 hours of receipt of
genicity have not been established for assessing efficacy. Field and other vaccine are brief, self-limited, and generalized and occur in association
CHAPTER 245  Active Immunizing Agents 2575

with fever. These seizures have not been demonstrated to result in the A single dose of Tdap is recommended universally for people 11
subsequent development of epilepsy or other neurologic sequelae. years or older, including adults, in place of the initial Td booster vaccine.
Predisposing factors include an underlying convulsive disorder, a personal The preferred schedule is to administer Tdap at the preventive visit for
history of previous convulsion, and a family history of convulsions.128 the 11- or 12- year-old children who have completed their primary
A small increase in the risk of febrile seizure but no increased risk in DTP/DTaP series.21,41,90 Adolescents and adults 11 years or older who
the development of epilepsy after immunization has been reported.627 received Td but not Tdap should receive a single dose of Tdap to provide
The incidence of local and febrile reactions after the administration protection against pertussis. Tdap can be administered regardless of
of acellular vaccines is significantly lower.231,541 Comparison of rates of the time since receipt of the last tetanus- or diphtheria-containing
adverse events with different acellular vaccines versus whole-cell vaccines vaccine.41 Adolescents between the ages of 11 and 18 years who have
demonstrated similar safety profiles for each of the vaccines.193,585 The never been vaccinated against tetanus, diphtheria, or pertussis should
most common adverse events include injection site reactions (e.g., initially receive a single dose of Tdap followed by Td for the subsequent
erythema, induration, tenderness) and mild system symptoms, including two doses at 4 weeks or more and 6 to 12 months later.
slight to moderate fever, drowsiness, irritability, and loss of appetite. No pertussis-containing vaccine is FDA approved for children between
Rates of local reactions increase with each subsequent dose of DTaP the ages of 7 and 10 years. If pertussis vaccine is not contraindicated,
vaccine.537,642 Booster doses of acellular pertussis vaccine may be associated children 7 through 10 years of age who are not fully vaccinated (i.e.,
with extensive local swelling (i.e., limb swelling), especially with vaccines have not received five doses of DTaP or four doses of DTaP if the fourth
having a high diphtheria content.554 The pathogenesis of this reaction dose was administered on or after the fourth birthday) should receive
is not understood, but it spontaneously resolves without sequelae and a single dose of Tdap to provide protection against pertussis. If additional
is not associated with an increased risk of similar adverse events occurring doses of tetanus and diphtheria toxoid–containing vaccines are needed
after receipt of the fifth dose.41 Severe reactions to acellular vaccines for children 7 through 10 years of age, Tdap vaccine is substituted as
such as prolonged crying of 3 hours or more, temperature greater than the first dose. The subsequent two doses should be Td vaccine at 4
or equal to 40.5°C (104.9°F), hypotonic-hyporesponsive episodes, and weeks or more and 6 to 12 months after the previous dose if indicated
seizures are rare.193,231,309,323,541,585,644 As with local and febrile reactions, for the catch-up series.29,90
their occurrence with acellular pertussis vaccination is significantly less Women should receive Tdap before becoming pregnant. Women
frequent than that after whole-cell vaccination. who have not been immunized previously with Tdap should receive
Syncope can occur after immunization with Tdap, is more common Tdap during pregnancy, preferably during the early third trimester (i.e.,
among adolescents and young adults, and can result in serious injury after 20 weeks’ gestation). If it is not administered during pregnancy,
if a vaccine recipient falls. Vaccinees should be seated and observed for Tdap should be administered in the immediate postpartum period.171,175
15 minutes after immunization. If syncope occurs, patients should be Additional immunization with Tdap should be administered in the
observed until symptoms resolve.41 early third trimester of each additional pregnancy. Every 10 years
When administered in the third trimester of pregnancy as recommended thereafter, booster vaccination should be provided by Td.
by the ACIP, there has been no association with an increased risk of As further protection against transmission of pertussis to infants
maternal or neonatal death, preeclampsia, hemorrhage, uterine rupture, who are at greatest risk for pertussis-related morbidity or mortality
caesarean section, renal failure, premature delivery, decreased birth weight, compared with other age groups, the cocoon strategy is recommended
or prolonged stay in a neonatal intensive care unit with administration to immunize mothers, families, and close contacts against pertussis to
of Tdap. Increases in poor birth outcomes or maternal adverse events decrease their likelihood of acquisition and subsequent transmission
have not been observed among women receiving multiple Tdap vaccines of B. pertussis to infants. Adolescents and adults (i.e., parents, siblings,
during pregnancies spaced less than 5 years apart.243,488,625 This finding grandparents, child care providers, and health care personnel) who
was confirmed in a review of reported adverse events following the recom- have or anticipate having close contact with an infant younger than 12
mendation that pregnant women received Tdap during the third trimester months should receive a single dose of Tdap to protect against pertussis
of each pregnancy regardless of prior vaccine history.489 if they have not previously received Tdap. Ideally, these people should
receive Tdap at least 2 weeks before beginning close contact with the
Serious Neurologic Illness infant.41,171
The National Childhood Encephalopathy Study (NCES), a large case-
control study from Great Britain that was published in 1985, estimated Contraindications and Precautions
that the occurrence of acute neurologic illness resulting in hospitalization The contraindications and precautions for administering pertussis
was 1 case per 140,000 DTP vaccinations.481 In a 10-year follow-up vaccine are based on adverse reactions associated with whole-cell vaccine.
study published in 1993, neurologic sequelae were found to be common Although reactions occurring after the administration of DTaP are much
occurrences, but no more so than in children with unrelated, acute less common than are those associated with DTP, the contraindications
neurologic illness in infancy,480 and reviews of the data have disputed and precautions for DTaP are the same.21,41,90 Adverse events temporally
the conclusion that pertussis vaccine can cause neurologic sequelae.14,128,622 related to pertussis immunization that contraindicate further administra-
Later data from Canada evaluating more than 12,000 pediatric hospi- tion of DTaP are as follows:
talizations for serious neurologic illness between 1993 and 2002 found • An immediate anaphylactic reaction. Subsequent immunization with
no association after administration of more than 6.5 million doses any of the three components of the vaccine should be avoided.
between development of encephalopathy and receipt of DTaP vaccina- • Encephalopathy occurring within 7 days. Children who experienced
tion.41 Taken together, the data do not support a role for whole-cell encephalopathy within 7 days after administration of a previous
pertussis vaccine in causing brain damage.115 dose of diphtheria and tetanus toxoids and pertussis vaccine (i.e.,
DTP, DTaP, or Tdap) not attributable to another identifiable cause
Indications should not receive additional doses of a vaccine that contains pertussis.
Vaccination against pertussis with DTaP is recommended routinely for Encephalopathy in this context is defined as a severe, acute, central
children at 2, 4, and 6 months of age, followed by a fourth dose at 12 nervous system disorder (e.g., coma, prolonged seizures) unexplained
to 18 months of age and a fifth dose at 4 to 6 years of age.21,41,90 Immuniza- by another cause and may manifest as major alterations in conscious-
tion can be started when the child is as young as 6 weeks of age if pertussis ness or generalized or focal seizures that persist for more than a few
is prevalent in the community. The interval between administrations hours without recovery within 24 hours. Additional doses of DT
of the three doses of the initial series can be as short as 4 weeks. The (or Td) vaccine should be substituted for any pertussis-containing
AAP and ACIP recommend exclusive use of acellular pertussis vaccines vaccine.
for all doses of the pertussis vaccine series.21,41,90 DTP is no longer available Postvaccination reactions constituting precautions are as follows:
in the United States. However, in many countries, including several in • Moderate or severe acute illness with or without a fever
Europe and developing countries, whole-cell vaccine remains the recom- • Guillain-Barré syndrome within 6 weeks after a previous dose of
mended product. tetanus toxoid–containing vaccine
2576 PART V  Prevention of Infectious Diseases

• A convulsion, with or without fever, occurring within 3 days of increasing incidence of antimicrobial-resistant pneumococci further
receiving DTP or DTaP vaccination underscores the need for developing effective pneumococcal vaccines
• Persistent, severe, inconsolable screaming or crying for 3 hours or for young children.88 Resistance of S. pneumoniae to multiple antibiotics
more within 48 hours of DTP or DTaP vaccination increased rapidly in the United States and even more rapidly in other
• Hypotonic-hyporesponsive episode occurring within 48 hours of parts of the world.381 Children younger than 2 years have the highest
DTP or DTaP vaccination rate of IPD but do not develop an effective antibody response to
• Temperature of 40.5°C (104.9°F) or greater that is unexplained by polysaccharide vaccine. Children 2 to 5 years of age may have relatively
another cause and occurs within 48 hours of DTP or DTaP poor responses to serotypes 6B, 14, 19F, and 23F, which are common
vaccination causes of pediatric infections and the most prevalent penicillin-resistant
With these adverse events occurring in temporal association with serotypes.
DTaP vaccination, the decision to administer additional doses of pertussis These factors prompted the development of conjugated polysac-
vaccine should be considered carefully. Children for whom vaccination charide–protein vaccines, two of which have been licensed in the United
is deferred because of moderate or severe acute illness should be vac- States. These vaccines are similar in design to the licensed Hib conjugate
cinated when their condition improves. In circumstances such as a vaccines. Because of the large number of serotypes of S. pneumoniae that
pertussis outbreak in which the potential benefits of pertussis immuniza- cause disease, development of these conjugate pneumococcal vaccines
tion outweigh the possible risks, vaccination is indicated, particularly was more difficult than the development of similar vaccines for Hib.
because these events have not been proved to cause permanent sequelae. Each pneumococcal antigen must be coupled to a protein carrier, and
The risk of these reactions occurring after receipt of DTaP is substantially the vaccine must be prepared to ensure that antigen is sufficient to induce
lower than after receipt of DTP. an immune response but insufficient to elicit an adverse reaction.
For children with an evolving neurologic disorder in the first year One potential problem with conjugate pneumococcal vaccines is
of life, pertussis immunization should be deferred until the nature and the need to immunize against many different serotypes of pneumococci.
cause of the disorder have been established. A personal history of having Because of local reactions to the protein component, conjugate vaccines
a stable neurologic condition (i.e., previous convulsion unrelated to that contain all implicated serotypes have proved difficult to produce.
DTaP vaccination, a history of seizure disorder, well-controlled seizures, Initial decisions for serotype inclusion need to be carefully planned for
or cerebral palsy) or a family history of convulsions is not a contraindica- specific groups. For instance, vaccine containing types 4, 6B, 9V, 14,
tion, and pertussis immunization should be administered on 18C, 19F, and 23F (i.e., PCV7) is necessary for prevention of otitis
schedule. media in the United States, whereas types 1, 2, and 5 need to be added
Certain conditions after DTaP vaccine, such as temperature of 40.5°C to prevent pneumonia in developing countries.
(104.9°F) or higher, collapse or shock-like state, persistent crying, or The use of conjugate vaccines against limited serotypes has led to
convulsions with or without fever, are a precaution to subsequent doses emergence of pneumococcal serotypes (e.g., 19A) that were previously
of DTaP. However, occurrence of one of these adverse reactions after less common but have proved equally concerning for IPD and emerging
DTaP vaccine in childhood is not a contraindication or precaution to resistance.102,161,364 Emergence of replacement serotypes led to the expan-
administration of Tdap to an adolescent or adult. A history of severe sion of the previous vaccine to the 13-valent pneumococcal conjugate
Arthus hypersensitivity reaction after a previous dose of a tetanus or vaccine (PCV13) in the United States.162 Routine infant immunization
diphtheria toxoid–containing vaccine administered less than 10 years using these conjugate vaccines led to significant reductions in the
earlier should lead to deferral of Tdap or Td immunization for 10 years instances of S. pneumoniae disease.263 Since the widespread introduction
after administration of the tetanus or diphtheria toxoid–containing of PCV7 in infants in 2000, rates of IPD have decreased markedly in
vaccine.21,41,90 the United States.263 The incidence of all IPDs has decreased by 80%
for children younger than 2 years and by up to 90% for infections
Pneumococcal Vaccine caused by vaccine and vaccine-related serotypes.42,161,686 An increase in
S. pneumoniae is a leading bacterial pathogen, especially among young nonvaccine serotype disease has occurred, but it is small compared
children, elderly people, and people with predisposing conditions. In with the overall reductions in vaccine-type disease.52,102,145,161
children, it is the most common cause of otitis media, occult bacteremia, A decline in the incidence of vaccine-type disease among unvaccinated
and bacterial pneumonia requiring hospitalization. After the widespread children and adults has occurred since the introduction of routine
introduction of conjugate Hib vaccination and subsequent marked vaccination for infants in the United States.145 Rates of IPD among
decline in occurrence of Hib meningitis, S. pneumoniae became a leading infants younger than 2 months have decreased significantly, providing
cause of bacterial meningitis in children in the United States. evidence that vaccinating children 2 to 23 months of age has led to
In some populations, such as Alaska Natives, the incidence of changes in pneumococcal carriage in infants who were too young to
bacteremia is markedly higher than that reported in other geographic receive PCV7.543 In surveillance of children with sickle cell disease from
areas of the United States.228 Groups considered at high risk for invasive 1998 to 2009, rates of IPD fell by 53%.528 As of 2012, PCVs are included
pneumococcal disease (IPD) include children with sickle cell disease, in national immunization programs in 86 of 194 WHO member states,
asplenia, Hodgkin disease, congenital humoral immunodeficiency, HIV with similar expectations of IPD decrease.174
infection, and nephrotic syndrome and recipients of organ transplants. Herd immunity is estimated to prevent twice as many cases as do the
Children who have received cochlear implants are at particular risk for direct effects of vaccination alone.145,161 Although the exact mechanism
developing meningitis.144,552 Other chronic diseases associated with an of herd immunity is uncertain, one hypothesis is that vaccinated children
increased risk of development of severe pneumococcal disease include are less likely to have nasal carriage of pneumococcus and therefore less
chronic cardiovascular and pulmonary diseases, diabetes mellitus, and pneumococcal transmission to their contacts.222 Use of conjugate vaccines
renal failure. The role of these chronic diseases in predisposing individuals has reduced nasopharyngeal carriage of vaccine serotypes with PCV7
to pneumococcal infection has been demonstrated primarily in adults. and PCV13.201,223,224 Pneumococcal conjugate vaccination also has led to
Mortality rates are highest among those who have bacteremia or increased nasopharyngeal colonization of children with nonvaccine
meningitis, elderly people, and patients with impaired humoral immunity serotypes.293,479 In surveillance of IPD cases among children younger than
or certain chronic diseases. 5 years, the introduction of PCV7 decreased penicillin-nonsusceptible
The purified polysaccharide vaccine has been effective in reducing cases by 64%, with serotypes not in PCV7 causing 78% to 97% of penicillin-
severe disease in the adult population,99 but it has little impact in young nonsusceptible IPD.331 This underscores the need for ongoing surveillance
children because the vaccine is not immunogenic in children younger of emerging nonvaccine serotypes with respect to nasopharyngeal coloniza-
than 2 years.42 The polysaccharide vaccine has not been effective in tion, antibiotic nonsusceptibility and of rates of IPD.613
preventing otitis media caused by S. pneumoniae.42 Several factors made
the development of new preventive strategies for pneumococcal disease Preparations
a high priority.212,214 Morbidity and mortality rates of pneumococcal Two pneumococcal vaccines are available for use in children in the
infection appear to be particularly high in developing countries. The United States. The first is a 13-valent vaccine (PCV13) licensed in 2010
CHAPTER 245  Active Immunizing Agents 2577

(Prevnar, Wyeth Vaccines), which contains the capsular polysaccharides was efficacious in preventing vaccine-type invasive disease in HIV-infected
from serotypes contained in the previous PCV7 (i.e., 4, 6B, 9V, 14, 18C, children.398 One of the trials demonstrated a reduction in the all-cause
19F, and 23F) and six additional serotypes (i.e., 1, 3, 5, 6A, 7F, and 19A) mortality rate for vaccine recipients.220 These data make a compelling
conjugated to mutant diphtheria toxin (CRM197).42 Serotypes included case for extending the benefits of pneumococcal conjugate vaccination
in PCV7 and potentially cross-reactive serotypes (i.e., 6A, 9A, 9L, 18B, to developing countries.
and 18F) accounted for 86% of cases of bacteremia, 83% of cases of Vaccination with purified polysaccharide vaccine results in serologic
meningitis, and 65% of AOM cases occurring in children younger than type-specific antibody in most healthy adults and older children.
6 years in the United States during 1978 to 1994.509 In 2007, the Active Immunocompromised patients may respond less well. In children younger
Bacterial Core surveillance estimated 4600 cases of IPD among children than 2 years, the antibody response is poor to most serotypes, including
younger than 5 years in the United States, with 2900 attributable to those most likely to cause infection, such as types 6A and 14.130,112,294
serotypes covered by PCV13 versus 60 cases covered by PCV7.161 The Patients with AIDS have impaired antibody responses to vaccination,
incidence of IPD from PCV13 or nonPCV7 serotypes was reduced by but asymptomatic HIV-infected adults do respond.130,112
64% and 93%, respectively, among children younger than 5 years by Vaccine efficacy in preventing serious pneumococcal infection has
2013, driven mostly by reductions of serotypes 19A and 7F.487 been demonstrated for the purified polysaccharide vaccine primarily
The second vaccine (i.e., PPV23 [Pneumovax]) is composed of in immunocompetent adults, including elderly people and those with
purified, capsular polysaccharide antigens of 23 pneumococcal serotypes. chronic diseases such as chronic pulmonary and cardiac disorders and
Although 90 different serotypes have been identified, vaccine serotypes diabetes mellitus, which predispose these patients to development of
in the 23-valent vaccine are historically responsible for 85% to 90% of pneumococcal infections. Efficacy against vaccine serotypes ranges from
adult infections and almost 100% of invasive disease and 85% of otitis 61% to 75% in adults.99,594 Investigations in adults in whom vaccine
media cases in children.112,294 Each vaccine is recommended in a dose protection against pneumococcal infection has been substantially less
of 0.5 mL that is given intramuscularly. have been criticized for methodologic problems.130,112 Efficacy in the
limited studies of children has been consistent with that for adults. In
Immunogenicity and Efficacy children with sickle cell disease or anatomic asplenia, an octavalent
The immunogenicity of the conjugate polysaccharide vaccine appears vaccine was highly effective in preventing bacteremic infection.51
to be determined by the pneumococcal polysaccharide serotype, rather
than by the carrier protein. Some serotypes (i.e., 14, 18C, and 19F) are Adverse Events
excellent immunogens because they elicit antibody protection after a Pneumococcal conjugate vaccines appear to be safe, and in 13 clinical
single dose, whereas others (i.e., 6B and 23F) require three doses of trials, the safety profile of PCV13 was comparable to that of PCV7.25,42,509
vaccine.259 Conjugate vaccine elicits immunologic memory.259 The The reactions most commonly reported have been local reactions at
antibody concentrations achieved after the initial series of three doses the injection site, but they occur at a lower frequency than do local
usually are sustained for only a few months and then decline to almost reactions with other childhood vaccines such as DTP.80
preimmunization levels. A dose of pneumococcal vaccine (i.e., polysac- Local reactions at the injection site, such as erythema and pain, are
charide or conjugate) given in the second year of life elicits an amnestic- reported for approximately 50% of recipients of the purified polysac-
type response. charide vaccine.42 However, more severe local and systemic reactions,
Prelicensure clinical efficacy of PCV7 was studied in a large, prospec- such as fever and myalgia, are rare events that occur in less than 1%
tive, placebo-controlled efficacy trial in northern California involving of vaccine recipients. Severe systemic reactions such as anaphylaxis
38,000 children. The vaccine was 89% effective in preventing invasive rarely have been reported.692 Preterm infants show safety results com-
disease caused by any pneumococcal serotype and 97% effective parable with those for term infants.463 In adults who were revaccinated
against disease caused by the seven vaccine serotypes.80 For noninvasive within 1 to 2 years in early studies, local reactions occurred more
disease, a decrease of 7% in the number of cases of otitis media and commonly than did those after initial immunization.130,112 However,
23% in doctor visits for recurrent otitis (i.e., six or more visits per year) subsequent investigations, including studies of children, indicated no
occurred for vaccinated children. The study also demonstrated an 11% increase in the incidence or severity of local or systemic reactions on
decrease in the number of clinical cases of pneumonia and efficacy of revaccination after longer intervals.401,557
26% against a first episode of radiograph-confirmed pneumonia in
vaccinees.333,600 Indications
Prelicensure immunogenicity of PCV13 was studied in a randomized, Recommendations for the use of PCV13 have been issued by the AAP
double-blind, active-controlled trial in which 663 US infants received and the ACIP.25,509 The AAP and ACIP recommend universal use of
at least one dose of PCV7 or PCV13. The study was powered for PCV13 in children 59 months or younger. For children to whom no
noninferiority of immunoglobulin antibody response measured by prior PCV7 or PCV13 has been administered before they reach 7 months
enzyme immunoassay and included subset analysis of functional antibody of age, four doses of PCV13 are recommended. Doses should be
responses measured by opsonophagocytosis assay (OPA).25,707 The administered routinely at 2, 4, 6, and 12 to 15 months of age. For
noninferiority criterion (i.e., immunoglobulin geometric mean concentra- children younger than 24 months who have not received their recom-
tions) was met for all shared serotypes; although slightly lower in the mended four doses of PCV7, a dose of PCV13 should be administered
PCV13 group, the functional OPA response was similar, suggesting at the next scheduled vaccination; the total number of doses is based
PCV13 has similar efficacy as PCV7 against shared serotypes. After a on the child’s age and described elsewhere.25,42,509
fourth dose of PCV13, the additional six serotypes also produced a A single supplemental dose of PCV13 is recommended for all children
functional OPA response in 97% to 100%. 14 months to 59 months who have previously received all four doses
The ability of PCV7 to protect children against AOM was evaluated of PCV7. A single dose of PCV13 is recommended for children 60
in an efficacy trial conducted in Finland.260 For prevention of AOM months to 18 years of age who are fully immunized with PCV7 and
caused by pneumococci of any serotype, efficacy was estimated to be have underlying medical conditions that increase their risk of IPD. For
34%, whereas efficacy against AOM irrespective of cause was 6%. Efficacy children 6 through 18 years of age, PCV13 should be given regardless
against AOM caused by vaccine-related serotypes was 57%, but an of previous PCV7 or PPSV23 administration.87 If not previously
increase of 33% in the rate of AOM episodes caused by nonvaccine administered, children 24 months to 18 years of age should receive the
serotypes occurred in the group receiving the heptavalent vaccine PPV23 to expand serotype coverage.
compared with controls. Despite of the increase in disease caused by The AAP recommends PPV23 for children 24 months or older and
nonvaccine serotypes, the net effect on pneumococcal AOM was a PCV13 for all children 59 months of age or older who belong to one
reduction of 34%. or more of the following risk groups:
RCTs enrolling African children and using a nine-valent pneumococcal • Immunocompetent children with chronic heart disease, chronic lung
conjugate vaccine demonstrated efficacy in the prevention of radiologi- disease, diabetes mellitus, cerebrospinal fluid leaks, or cochlear
cally confirmed pneumonia and vaccine-type IPD.220,398 The vaccine implants
2578 PART V  Prevention of Infectious Diseases

• Children with functional or anatomic asplenia, including sickle cell The elimination of poliovirus infection has been achieved primarily
disease or other hemoglobinopathies, congenital or acquired asplenia, through the use of OPV (i.e., Sabin strain). This product had been the
or splenic dysfunction vaccine of choice for children in the United States since the early 1960s
• Children with immunocompromising conditions, including HIV because it induced optimal intestinal immunity, was painless to
infection; chronic renal failure and nephrotic syndrome; diseases administer, and secondarily immunized some contacts by fecal-oral
associated with treatment with immunosuppressive drugs or radiation spread of the vaccine virus and contributed to the immunity of the
therapy, including malignant neoplasms, leukemias, lymphomas, population.105 Because 8 to 10 cases of vaccine-associated paralytic polio
and Hodgkin disease; or solid organ transplantation and congenital occurred annually and the risk of exposure to wild poliovirus had been
immunodeficiency markedly reduced or eliminated in the United States, expanded use of
For previously vaccinated children 18 years or younger who are at IPV (i.e., Salk strain) was recommended by the CDC and the AAP
high risk for severe pneumococcal infection, revaccination with PPV23 beginning in 1997.16,129
after 5 years is recommended.25,87,509 For children between the ages of In 1999, as a result of progress in the global eradication of polio-
6 and 18 years who have not received PCV13 but have received at myelitis, the need for further reduction in the risk of acquiring vaccine-
least one PPSV23 dose, a single PCV13 dose should be given at least associated paralytic polio, and the acceptance of IPV by parents and
8 weeks after the last PPSV23 dose, regardless of previously given physicians,131 IPV was recommended for the first two doses of poliovirus
PCV7.176 This includes children who have functional (e.g., sickle cell vaccine for routine childhood vaccination.18,134 To eliminate the risk of
disease) or anatomic asplenia, children and adolescents with HIV vaccine-associated paralytic polio completely, an all-IPV schedule was
infection, and those who have a rapid antibody decline (i.e., nephrotic recommended in January of 2000 for routine childhood vaccination
syndrome, renal failure, or organ transplantation).295 Revaccination with in the United States.19,137,134 IPV is the only vaccine used in developed
PPV23 should be considered for high-risk older children and adults countries.
who were vaccinated 6 years or more earlier, but no more than two Since 2000, there have been ongoing outbreaks of paralytic polio-
total PPV23 doses are necessary. The interval between polysaccharide myelitis caused by circulating VDPVs that are neurovirulent polioviruses
and conjugate vaccines should be at least 8 weeks, regardless of the derived from the Sabin OPV vaccine strain. The epidemics have occurred
order given.399 among underimmunized children living in certain economically deprived
regions. The low immunization rates in these areas have permitted
Contraindications these viruses to circulate for long periods and, by continuous mutation,
No contraindications to initial vaccination exist.25,509 The safety of to acquire biologic properties that are indistinguishable from naturally
pneumococcal vaccine in pregnant women has not been evaluated, but occurring wild polioviruses. Interruptions in OPV administration and
adverse consequences for the fetus have not been observed in neonates the previous eradication of the corresponding serotype of indigenous
whose mothers were vaccinated inadvertently during pregnancy. Ideally, wild poliovirus were the critical risk factors for all VDPV outbreaks.387
women at high risk for pneumococcal disease should be vaccinated Circulation of VDPVs underscores the critical importance of eliminating
before pregnancy. For people who have had a severe reaction, such as the last pockets of wild poliovirus, maintaining universally high levels
anaphylaxis or a localized, severe hypersensitivity response, revaccination of polio vaccine coverage (>90% of young children), withdrawing all
should be avoided. OPVs as soon as wild poliovirus has been eradicated globally, and
continuing poliovirus surveillance into the foreseeable future.
Poliovirus Vaccine
The widespread implementation of poliovirus vaccine programs resulted Preparations
in a dramatic reduction in the incidence of paralytic poliomyelitis Trivalent IPV is the only vaccine available for routine infant and child-
throughout the world. In contrast to the prevaccine era, when more hood immunization in the United States. IPV is prepared by inactivation
than 18,000 cases of paralytic disease occurred in the United States of naturally occurring polioviruses by treatment with formaldehyde.
annually, the last known case in this country caused by indigenous Five trivalent IPV vaccines are available in the United States: IPOL
wild-type virus occurred in 1979.178 Other than rare imported cases, (single-component vaccine [Sanofi Pasteur]), Pediarix (in combination
the only cases of paralytic poliomyelitis in the United States since then with DTaP vaccine and HBV vaccine [GlaxoSmithKline]), Pentacel (in
have been vaccine related. combination with DTaP and Hib vaccines [Sanofi Pasteur]), Quadracel
The effectiveness of polio vaccination led the WHO to launch the (in combination with DTaP vaccine [Sanofi Pasteur]), and Kinrix (in
Global Polio Eradication Initiative in 1988, when approximately 350,000 combination with DTaP vaccine [GlaxoSmithKline]). These vaccines
people succumbed to polio each year. The initiative initially made rapid are produced in monkey kidney (Vero) cells except for Pentacel, which
gains; the number of new wild poliovirus cases decreased by 99% between is grown in human diploid cells, and each contains 40, 8, and 32 D-antigen
1988 and 2005. However, ongoing transmission threatened the success units of poliovirus types 1, 2, and 3, respectively. The single-component
of the polio eradication program, and the Polio Eradication and Endgame vaccine is the only IPV preparation that contains preservative.
Strategic Plan 2013–18 was developed under the auspices of the WHO The trivalent OPV formulations contain approximately 106.5 50%
to eradicate wild poliovirus, vaccine-associated paralytic polio (VAPP), tissue culture infectious doses (TCID50), 105.5 TCID50, and 106.2 TCID50
and circulating vaccine-derived poliovirus (VDPV).701 The phased plan of poliovirus types 1, 2, and 3, respectively, in a 10 : 1 : 3 ratio. The
was implemented in places where OPV was used routinely. At least one unequal contribution of each type to the trivalent preparation represents
dose of inactivated polio vaccine (IPV) was introduced in 2015, and a balanced formulation designed to account for the more efficient
bivalent OPV (bOPV, types 1 and 3) replaced trivalent OPV (tOPV) replication of type 2 OPV virus in the gastrointestinal tract and to
starting in 2016. The type 2 component of tOPV is being removed avoid interference with the replication of the two latter types. The live
because it causes more than 90% of all cVDPV cases and approximately oral OPV strains are grown in Vero cells and do not contain a preservative.
40% of vaccine-associated paralytic polio cases, it interferes with the OPV is no longer available in the United States.
immune response to the other two types, and wild poliovirus type 2 Monovalent OPV (mOPV) vaccines (types 1 and 3) are recommended
has been eradicated since 1999. for use in supplementary immunization campaigns where only wild
As of 2016, four of the six regions of the WHO have been certified poliovirus type 1 or type 3 alone is circulating, with the purpose of
polio-free: the Americas (1994), Western Pacific (2000), Europe (2002), controlling outbreaks. Two doses of mOPV administered within
and South East Asia, including India (2014). Eighty percent of the world’s 2 weeks is used as part of the short-interval additional dose approach,
population currently lives in polio-free areas. However, Afghanistan, which is intended to rapidly boost population immunity in certain
Nigeria, and Pakistan are the three remaining reservoirs of endemic settings.670
disease, largely because regional conflicts and poor health care infra-
structures hinder widespread immunization campaigns. Countries with Immunogenicity and Efficacy
ongoing outbreaks of cVDPV include Guinea, Lao People’s Democratic Neutralizing antibodies are detectable to all three poliovirus types in
Republic, Madagascar, Myanmar, Nigeria, and Ukraine.516,539,701 99% of recipients of IPV after two doses and in 100% after the third
CHAPTER 245  Active Immunizing Agents 2579

dose.468,606 Detectable antibody persists at protective levels for at least childhood. However, vaccination is recommended for individuals who
5 years, although geometric mean titers decline considerably.630 A have an increased risk of exposure, including people traveling to countries
meta-analysis revealed that vaccine efficacy of the currently licensed where poliomyelitis is epidemic or endemic, members of communities
enhanced-potency IPV formulations ranges from 33% to 47% after of specific population groups experiencing wild poliovirus disease, health
one dose and 79% to 90% after two doses in infants older than 10 care workers in close contact with patients who may be excreting wild
weeks.308 poliovirus, and laboratory workers in contact with specimens that may
An optimal immune response to trivalent OPV requires multiple contain wild-type poliovirus. Previously immunized adults who are at
doses. In developed countries, three doses given at least 2 months apart increased risk for exposure to poliomyelitis, such as those traveling to
are sufficient, with antibody prevalence to all three types being approxi- countries where poliomyelitis is still endemic, should receive a single
mately 96% after the third dose.468 Detectable serum antibody to all dose of IPV. Available data do not indicate the need for more than a
three types persists in 84% to 98% of vaccinees 5 years after receiving single lifetime booster dose of IPV. Adults who are unvaccinated or
primary immunization.411 whose vaccination status is not documented should receive a primary
OPV efficacy was directly evaluated during a type 1 poliovirus vaccination series with IPV. This consists of two doses of IPV at 4- to
outbreak in Taiwan in the early 1980s. During this outbreak, vaccine 8-week intervals and a third dose 6 to 12 months after the second dose.
efficacy was estimated to be 82%, 96%, and 98% for one, two, and three
or more doses, respectively.390 In tropical countries, the series of OPV Precautions and Contraindications
at 6, 10, and 14 weeks of age recommended by the WHO EPI fails to IPV is contraindicated in people who have ha an anaphylactic reaction
produce active immunity in a significant proportion of infants. Low after receiving a previous dose of IPV or an anaphylactic reaction to
seroconversion rates have been documented in many locations, averaging one of the antibiotics in the vaccine preparation (i.e., streptomycin,
73%, 90%, and 70% for types 1, 2, and 3, respectively.526 Diarrheal polymyxin B, or neomycin).19,43,134 Poliomyelitis vaccination usually is
disease at the time of immunization is a major factor.496 The impact of contraindicated in pregnant women because of the theoretical risk of
diarrhea on seroconversion persists despite the administration of three harm to the fetus. However, no deleterious effects from IPV administered
or four OPV doses. during pregnancy have been demonstrated; and if immediate protection
The monovalent OPV vaccines have higher type-specific seroconver- against poliomyelitis is needed, IPV may be given.
sion rates than does the trivalent vaccine and are considered more
effective when used in response to an outbreak caused by one poliovirus Rotavirus Vaccine
type.103 Rotavirus infection is the most common cause of severe diarrhea disease
in infants and young children worldwide and continues to have a major
Adverse Events global impact on childhood morbidity and mortality, causing an estimated
No serious adverse events have been associated with use of the currently 527,000 deaths of children younger than 5 years each year or approximately
available IPV vaccine.19,43,134 Because IPV vaccine contains trace amounts 5% of all childhood deaths worldwide.521 In the United States before the
of streptomycin, neomycin, and polymyxin B, allergic reactions are introduction of vaccine in 2006, an estimated 3 million rotavirus infections
possible in recipients with hypersensitivity to one or more of these occurred every year, and 95% of children had at least one rotavirus
antibiotics. infection by 5 years of age. Rotavirus infection was responsible for more
The OPV vaccine can cause vaccine-associated paralytic polio, the than 400,000 physician visits, more than 200,000 emergency department
overall risk for which is approximately one case per 2.4 million doses (ED) visits, 55,000 to 70,000 hospitalizations, and 20 to 60 deaths each
distributed. The rate after the first dose is approximately one case per year of children younger than 5 years.298,688 In the prevaccine era, rotavirus
750,000 doses, including vaccine recipient and contact cases. Immu- accounted for 30% to 50% of all hospitalizations for gastroenteritis
nodeficiency-related VDPV excretion by people with primary immu- among US children younger than 5 years.217
nodeficiencies is a rare complication of OPV use and is associated with Rotavirus gastroenteritis is preventable with live oral rotavirus
chronic excretion and fatal disease.98 vaccines. In 1998, the only approved rotavirus vaccine, a tetravalent
rhesus-based rotavirus vaccine (RRV-TV) (Rotashield, Wyeth Labora-
Indications tories), was withdrawn from the market in the United States because
The polio vaccination series in the United States consists of four doses of an association with intussusception among vaccinated infants.495 In
of IPV administered subcutaneously or intramuscularly.19,43,134 The first 2006, a bovine-based pentavalent rotavirus vaccine (RotaTeq, Merck &
and second doses are administered at 2 and 4 months of age, respectively. Co.) was licensed by the FDA for use in infants in the United States.
The third dose is usually given at 6 to 18 months of age, and a fourth The ACIP of the CDC and the AAP have recommended universal
dose is given routinely at 4 to 6 years of age, before school entry. vaccination of US infants against rotavirus.24,217 In 2008, a live attenuated
Administration of a fourth dose is not necessary if the third dose was human rotavirus vaccine (Rotarix, GlaxoSmithKline) was licensed in
given on or after the child’s fourth birthday. IPV is a component of the United States.
Pediarix, Pentacel, Quadracel, and Kinrix. Pediarix may be given from The epidemiology of rotavirus disease in the United States has changed
6 weeks through 6 years of age. Pentacel may be used 6 weeks through dramatically since rotavirus vaccines became available in 2006. National
4 years of age, and Quadracel and Kinrix may be used for children 4 declines in rotavirus detection have been reported, ranging from 58%
to 6 years of age. to 90% in each of the seven postvaccine years compared with all seven
The WHO continues to recommend OPV as the vaccine of choice prevaccine years combined. A biennial pattern of rotavirus activity
for global eradication of wild poliovirus in endemic areas and during emerged in the postvaccine era, with years of low activity and highly
epidemics. Since 2015, at least one additional dose of IPV is recommended erratic seasonality alternating with years of moderately increased activity
from 14 weeks of age, with or without a concurrent OPV dose, in and seasonality similar to that seen in the prevaccine era.9 Other US
preparation for withdrawing all OPV after global eradication has been studies show the large public health impact of routine rotavirus vaccination
certified. Since 2016, trivalent OPV has been switched to bivalent OPV in reducing the circulation of rotavirus among US children. Declines in
(i.e., types 1 and 3). In the immunization schedule of the WHO EPI, laboratory-confirmed rotavirus hospitalization632 and reductions in
doses of OPV are recommended at birth and when the child is 6, 10, outpatient visits, emergency room visits, acute gastroenteritis, and
and 14 weeks of age.29 In geographic areas with endemic polio, a dose rotavirus-coded hospitalizations have been reported.215 From 2007 through
may be given when the neonate is discharged from the hospital (i.e., 2011, more than 176,000 hospitalizations, 242,000 emergency department
the zero dose). Supplementary doses often are given during mass com- visits, and 1.1 million outpatients visits due to diarrhea were averted,
munity programs in these areas. Breastfeeding does not interfere with resulting in costs savings of $924 million over the 4-year period.427
successful immunization with OPV.
Routine poliovirus vaccination is not necessary in adults residing Preparations
in the United States. These individuals are at minimal risk for exposure, Two rotavirus vaccines are licensed for use in the United States. The
and most are adequately protected because of vaccination during pentavalent rotavirus vaccine (RV5) is an oral vaccine that contains
2580 PART V  Prevention of Infectious Diseases

five live reassortant rotaviruses. The rotavirus parent strains of the observed in the RV5 group versus five cases of intussusception in the
reassortants were isolated from human and bovine hosts. Four reassortant placebo group (multiplicity-adjusted relative risk, 1.6). The data did
rotaviruses express one of the outer capsid proteins (i.e., G1, G2, G3, not suggest an increased risk of intussusception relative to placebo.
or G4) from the human rotavirus parent strain and the attachment Among vaccine recipients, no confirmed cases of intussusception occurred
protein (i.e., P7[5]) from the bovine rotavirus parent strain. The fifth within the 42-day period after administration of the first dose, which
reassortant virus expresses the attachment protein (i.e., P1A[8]) from was the period of highest risk for the previously licensed RRV-TV vaccine.
the human rotavirus parent strain and the outer capsid protein (i.e., No evidence of clustering of cases of intussusception was observed
G6) from the bovine rotavirus parent strain. The reassortants are within a 7- or 14-day window after immunization for any dose. For
propagated in Vero cells using standard tissue culture techniques. the 1-year follow-up period after administration of the first dose, 13
RV5 vaccine is provided in a squeezable plastic dosing tube with a cases of intussusception were observed in the RV5 group versus 15
twist-off cap designed to allow for the vaccine to be administered directly cases of intussusception in the placebo group (multiplicity-adjusted
to infants by mouth. Each tube contains a single 2-mL dose of the relative risk = 0.9).
vaccine as a buffered-stabilized liquid solution. Some studies performed outside the United States detected a low
The human rotavirus vaccine (RV1) is a G1 P1A[8] virus attenuated level of increased risk of intussusception after rotavirus immunization
by passage in cell culture. RV1 is provided as a lyophilized powder that shortly after the first dose. The level of risk observed in these postmarket-
is reconstituted with a supplied diluent before administration. ing studies is substantially lower than the risk of intussusception after
Both rotavirus vaccines must be stored at refrigerator temperatures immunization with RotaShield, the previous rotavirus vaccine.100,524,655
(2°C–8°C [35°F–46°F]) and protected from light. RV1 diluent may be US postmarketing studies have also identified a small increased risk of
stored at room temperature. The vaccines must not be frozen. The shelf intussusception with RV5 and RV1, usually within a week after the first
life of properly stored vaccine is 24 months. RV5 should be administered or second vaccine dose. This additional risk is estimated to range from
as soon as possible after being removed from refrigeration. RV1 should about one case per 20,000 to 100,000 US infants who receive rotavirus
be administered within 24 hours of reconstitution. Reconstituted RV1 vaccine.73,325,601,679,708
may be stored at refrigerator or room temperature. Both vaccines are well tolerated and have a low reactogenicity profile
No studies have addressed the interchangeability of the rotavirus when given alone. They do not cause clinically significant increases in
vaccines. reactogenicity when coadministered with other routine childhood
vaccines.234,563 For both vaccines, the incidence of fever, vomiting, diarrhea,
Immunogenicity and Efficacy and irritability were measured in the clinical trials. For RV5, no significant
The immune correlates of protection from rotavirus infection and disease difference versus placebo was observed in the incidence of fever or
are not fully understood. In a large phase III clinical trial of RV5, an severe fever and irritability or severe irritability. A 3% increase in the
increase in titer of rotavirus group-specific serum IgA antibodies was incidence of diarrhea and vomiting was observed with RV5, but these
used as one of the measures of the immunogenicity. Serum samples symptoms were mild and did not require treatment.83,236,660 For RV1,
were obtained from a subset of study participants before they were no difference versus placebo was observed in the incidence of diarrhea,
immunized and approximately 2 weeks after they received the third fever or severe fever, vomiting or severe vomiting, and irritability or
dose. Seroconversion was defined as a threefold or greater increase in severe irritability within 14 days of immunization with any dose.235,535,657,659
antibody titer from baseline. Seroconversion rates for IgA antibody to There was no statistically significant increased risk of death or other
rotavirus were 95% among vaccine recipients and 14% among recipients serious adverse events with either vaccine compared with placebo.
of the placebo.660 Vaccine virus shedding in stool has been documented for both
The efficacy of RV5 was evaluated in two phase III trials.83,660 In rotavirus vaccines. Rotavirus shedding occurs in approximately 9% of
these trials, the efficacy of RV5 after completion of a three-dose regimen RV5 recipients after the first dose but rarely after subsequent doses.236
against rotavirus gastroenteritis of any severity was 74%, and against For RV1, live rotavirus shedding in stool occurs in approximately 25%
severe rotavirus gastroenteritis, it was 98 percent. Efficacy was observed of recipients, with peak excretion occurring about day 7 after the first
against all G1 to G4 and G9 serotypes, but relatively few non-G1 rotavirus dose. With the use of more sensitive real-time PCR tests for rotavirus
cases were reported. RV5 reduced the incidence of office visits by 86%, shedding, detection rates for viral shedding were demonstrated in 20%
emergency department visits by 94%, and hospitalizations for rotavirus to 30% in recipients of RV5 and in 80% to 90% of those receiving
gastroenteritis by 96%. The efficacy of RV5 in the second rotavirus RV1.355,611 Both vaccines have been transmitted from vaccine recipients
season after immunization was 63% against rotavirus gastroenteritis to unvaccinated siblings, but no significant gastrointestinal symptoms
of any severity and 88% against severe rotavirus gastroenteritis. were seen in the unvaccinated sibling after transmission.527,559 Shedding
Neither breastfeeding nor concurrent administration of other child- and transmission are not considered significant safety concerns because
hood vaccines appears to diminish the efficacy of a three-dose series of the attenuated nature of the rotavirus vaccine strains and the lack
of RV5. The efficacy of RV5 between doses of a three-dose series and of severe gastrointestinal symptoms after transmission.
with less than three doses (i.e., incomplete regimen) was explored in In 2010, porcine circovirus or porcine circovirus DNA was detected
post hoc analyses of the large efficacy study of RV5. The vaccine reduced in both rotavirus vaccines.68,297,413,469 There is no evidence that this virus
the rates of combined hospitalizations and ED visits for G1 through is a safety risk or causes illness in humans.412
G4 rotavirus gastroenteritis by 100% between doses 1 and 2 and 91%
between doses 2 and 3. RV5 reduced the rates of rotavirus gastroenteritis Indications
regardless of serotype by 82% between doses 1 and 2 and 84% between Infants should receive three doses of RV5 administered orally at 2, 4,
doses 2 and 3.237 and 6 months of age or two doses of the RV1 at 2 and 4 months of
Phase III clinical trials of RV1 efficacy have involved more than age.24,217 The first dose should be administered when the child is between
21,000 infants 6 through 12 weeks of age, primarily in two studies in 6 and 14 weeks of age (i.e., on or before 14 weeks, 0 days of age).
Latin America and Europe.569,658 After completion of a two-dose RV1 Subsequent doses should be administered at a minimum of 4-week
regimen, the efficacy of rotavirus vaccine against severe rotavirus intervals, and all doses of vaccine should be administered by the time
gastroenteritis (i.e., Latin American study) was 85%, and against any the child is 32 weeks of age (i.e., on or before 32 weeks, 0 days).
rotavirus gastroenteritis (i.e., European study), it was 87%. RV1 reduced Immunization should not be initiated for infants older than 14
hospitalization for rotavirus gastroenteritis by 85% to 100%, depending weeks because of insufficient data on the safety of the first dose of
on the study. The efficacy of fewer than two doses is not known. rotavirus vaccine in older infants. Vaccine should not be administered
after 8 months of age because of insufficient data on the safety and
Adverse Events efficacy of rotavirus vaccine in infants after this age. For infants for
Safety with respect to intussusception was evaluated for 71,725 subjects whom the first dose of rotavirus vaccine is inadvertently administered
enrolled in phase III efficacy trials of RV5.83,660 For the prespecified off-label at 15 weeks or older, the rest of the rotavirus immunization
42-day postimmunization end point, six cases of intussusception were series should be completed according to the schedule defined earlier
CHAPTER 245  Active Immunizing Agents 2581

because timing of the first dose should not affect the safety and efficacy congenital malabsorption syndromes, Hirschsprung disease, short-gut
of the second and third doses. syndrome, persistent vomiting of unknown cause).
The rotavirus vaccine series should be completed with the same A history of intussusception is a contraindication for the use of
product whenever possible. However, vaccination should not be deferred both rotavirus vaccines.165 Some data suggest that infants with a history
if the product used for a prior dose or doses is not available or is not of intussusception may be at higher risk for a repeat episode than other
known. In this situation, the provider should continue or complete the infants.
series with the product that is available. If any dose in the series was
RV5 (RotaTeq) or the vaccine brand used for any prior dose in the Rubella Vaccine
series is not known, a total of three doses of rotavirus vaccine should Rubella is a viral disease that usually manifests as a mild febrile rash
be administered. Infants documented to have had rotavirus gastroenteritis illness in adults and children; however, 20% to 50% of infected people
before receiving the full course of rotavirus immunizations should still are asymptomatic. Rubella can have severe adverse effects on the fetuses
start or complete the three-dose schedule because the initial infection of pregnant women who contract the disease during the first trimester
frequently provides only partial immunity. of pregnancy. It causes a wide range of congenital defects known as
Infants who are being breastfed can receive rotavirus vaccine. Like congenital rubella syndrome (CRS). The primary objective of the rubella
other childhood vaccines, rotavirus vaccine can be administered to vaccination program is to prevent intrauterine rubella infection. The
infants with transient, mild illnesses, with or without low-grade fever.408 primary strategies for rubella control in the United States are universal
Rotavirus vaccine can be administered together with DTaP, Hib, IPV, childhood vaccination, prenatal screening of pregnant women for rubella
hepatitis B, and pneumococcal conjugate vaccines. immunity, and vaccination of rubella-susceptible women after
The AAP and ACIP support immunization of preterm infants under delivery.
the following conditions. The infant is at least 6 weeks of age, the infant In the prevaccine era, epidemics of rubella occurred every 6 to 9
is clinically stable, and the first dose of vaccine is given at the time of years, with the last major epidemic in the United States taking place in
discharge or after the infant has been discharged from the hospital 1964 through 1965.694 The incidence of reported cases of rubella fell
nursery. sharply after routine rubella immunization of young children was initiated
Infants living in households with people who have or are suspected in the United States in 1969. From the estimated 2 million cases per
of having an immunodeficiency disorder or impaired immune status year in the prevaccine era, fewer than 1000 cases were reported in 1983.
or with a pregnant woman can be immunized. To minimize potential During 1989 through 1991, a resurgence of rubella occurred, primarily
virus transmission, all members of the household should employ because of outbreaks among unvaccinated adolescents and young adults
measures such as good handwashing after contact with the feces of the who initially were not recommended for vaccination and in religious
immunized infant (e.g., after changing a diaper) for at least 1 week communities with low rubella vaccination coverage.107 As a result of
after the first dose of rotavirus vaccine has been given. the rubella outbreaks, two clusters of approximately 20 CRS cases
An infant who regurgitates, spits out, or vomits during or after occurred.424,475 Outbreaks during the mid-1990s occurred in settings
receiving a dose of rotavirus vaccine should not have that dose re- where young adults congregated and involved unvaccinated people who
administered. The infant can receive the remaining recommended doses belonged to specific racial or ethnic groups.121 Further declines occurred
of rotavirus vaccine at appropriate intervals. If a recently immunized as rubella vaccination efforts increased in other countries in the WHO
child is hospitalized for any reason, no precautions other than standard Region of the Americas. From 2001 through 2004, reported rubella and
precautions need be taken to prevent the spread of vaccine virus in the CRS cases were at an all-time low, with an average of 14 reported rubella
hospital setting. cases a year, four CRS cases, and one rubella outbreak (defined as three
or more cases linked in time or place).550
Contraindications and Precautions In October 2004, 35 years after initiation of the rubella immunization
Rotavirus vaccine should not be administered to infants who have severe program, an independent panel of international experts was convened
hypersensitivity to any component of the vaccine or individuals who by the CDC to assess progress toward elimination of rubella and CRS.
have experienced a serious allergic reaction to a previous dose of rotavirus Based on data showing fewer than 25 cases of rubella reported each
vaccine.24,217 The RV1 oral applicator contains latex rubber. RV5 may year since 2001, at least 95% vaccination coverage among school-aged
be preferred for children at high risk for latex sensitization. children, an estimated 91% population immunity, adequate surveillance
Practitioners should consider the potential risks and benefits of to detect rubella outbreaks, and a pattern of virus genotypes consistent
administering rotavirus vaccine to infants with known or suspected with virus originating in other parts of the world, panel members
altered immunocompetence. Children and adults who are immuno- concluded unanimously that rubella was no longer is endemic in the
compromised because of congenital immunodeficiency, bone marrow United States.549
transplantation, or solid organ transplantation sometimes experience Rubella continues to be endemic in many parts of the world.
severe, prolonged, and even fatal rotavirus gastroenteritis. However, Internationally imported rubella cases may give rise to indigenous
no safety or efficacy data are available for the administration of transmission. From 2005 through 2011, a median of 11 rubella cases
rotavirus vaccine to infants who are potentially immunocompromised was reported each year in the United States (range, 4–18). Two rubella
with the exception of infants with severe combined immune outbreaks involving three cases and four total CRS cases were reported.
deficiency (SCID). SCID is a contraindication for the use of both rotavirus Among the 67 rubella cases reported from 2005 through 2011, a total
vaccines.157 Gastroenteritis, including severe diarrhea and prolonged of 28 cases (42%) were known importations.551 In 2011, an expert panel
shedding of vaccine virus, has been reported in infants who were reviewed available data and unanimously agreed that rubella elimination
administered live oral rotavirus vaccines and later identified as having had been maintained in the United States.520
SCID.61 From 2012 through 2015 a total of 29 rubella cases (range, 5–9
Rotavirus vaccine should not be administered to infants with acute, cases/year) and 6 CRS cases (range, 1–3 cases/year) have been reported
moderate to severe gastroenteritis until the condition improves. Rotavirus in the United States. The United States needs to continue its vigilance
vaccine has not been studied among infants with concurrent acute against rubella and CRS by maintaining high vaccination rates among
gastroenteritis, in whom its immunogenicity and efficacy theoretically children; ensuring vaccination among women of childbearing age,
can be compromised. Infants with moderate to severe illness should especially women born outside the United States; continuing surveillance
be immunized as soon as they have recovered from the acute phase of of rubella and CRS; and responding rapidly to any outbreak.146
the illness.
Infants with preexisting chronic gastrointestinal conditions and who Preparations
are not undergoing immunosuppressive therapy should benefit from Since 1979, RA 27/3 (Rubella abortus, 27th specimen, third extract)
rotavirus vaccine immunization, and the benefits outweigh the theoretical vaccine, prepared in human diploid tissue culture, has been the only
risks. However, the safety and efficacy of rotavirus vaccine have vaccine available in the United States; it replaced the earlier HPV-77
not been established for infants with these preexisting conditions (e.g., and Cendehill vaccines. RA 27/3 induces higher antibody titers and
2582 PART V  Prevention of Infectious Diseases

more closely parallels the immune response after natural infection than history of having clinical rubella, unless serologic tests confirm
did previous vaccines.314,420,425,515,676 Two rubella-containing vaccines, immunity.
MMR and MMRV, are available. Both vaccines should be kept at 2°C Emphasis should be placed on the immunization of the postpubertal
to 8°C (35.6°F–46.4°F) or colder during storage and should be protected male and female population, especially college students and those in
from light to avoid inactivation of the virus. After reconstitution, the the military. Rubella vaccine also should be administered to adolescent
vaccine should be used within 8 hours. girls and women of childbearing age who lack a history of previous
vaccination. Other opportunities for immunization include premarital
Immunogenicity and Efficacy screening, routine gynecologic examinations, visits for neonatal and
Immunization with rubella vaccine induces humoral and cell-mediated well-child care, or other medical visits. The immediate postpartum
immunity. At least 95% of susceptible vaccinees 12 months or older period is an excellent time for giving immunizations. Rubella vaccine
develop antibody titers that are protective after a single dose of can be given after administration of anti-Rho(D) immune globulin,
vaccine.275,281,288,607,639,676 After a second dose of rubella vaccine, approxi- but serologic testing to determine whether seroconversion has occurred
mately 99% had detectable rubella antibody and approximately 60% should be performed at least 8 weeks after vaccination. When practical,
had a fourfold increase in titer.372,404,423 potential vaccinees may be screened for susceptibility. However, vaccina-
Follow-up studies indicate that one dose of rubella vaccine can tion of girls and women of childbearing age is justifiable and may be
provide long-lasting immunity. Vaccine-induced rubella antibodies have preferable, without previous serologic testing of women not known to
persisted in more than 90% of vaccinees 16 years after receiving a single be pregnant.
dose of vaccine, but antibody levels decreased over time.190,345,353,421,540 Adults in the United States who were born in countries where rubella
Although levels of vaccine-induced rubella antibodies can decrease over vaccination was not offered are at higher risk for contracting rubella
time, data from surveillance of rubella and CRS suggest that waning and having infants with CRS. Health care practitioners who treat foreign-
immunity with increased susceptibility to rubella disease does not occur. born adults should document the rubella immunity of these patients
Among people receiving two doses, approximately 91% to 100% had with a written record of rubella-containing vaccine or by serologic
detectable antibodies 12 to 15 years after receiving the second dose.227,423 testing. Susceptible adults, especially women of childbearing age, should
Outbreaks of rubella in vaccinated populations are rare. Studies be vaccinated. During rubella outbreaks, all susceptible people who
demonstrate rubella vaccine is approximately 97% effective in preventing have no contraindications to rubella vaccine should be identified and
clinical disease after a single dose (range, 94–100%).69,225,283 vaccinated.
Lifelong protection against clinical reinfection or asymptomatic
viremia, or both, usually results from a single dose of vaccine given Precautions and Contraindications
early in childhood. In some cases, vaccinees exposed to natural rubella Specific contraindications to the administration of live rubella vaccine
developed a rise in antibody titer unassociated with clinical symptoms. include pregnancy; severe febrile illness; known history of anaphylactic
Reinfection is associated only rarely with viremia. Significant pharyngeal reaction to rubella vaccine, gelatin, or neomycin, which are contained
shedding also is observed infrequently. Person-to-person transmission, in the vaccine; and immunodeficiency conditions (i.e., malignancy,
however, has not been reported. Reinfection caused by wild-type rubella primary immunodeficiency disease, immunosuppressive or corticosteroid
virus also may be observed in individuals with previous natural rubella therapy, and radiation therapy).44,471 People with mild immunosuppres-
infection. The risk of CRS developing from rubella reinfection during sion, such as those with asymptomatic HIV infection or those taking
pregnancy is extremely low.561 short-term or low-dose corticosteroids, may be vaccinated.
Postpubertal women of childbearing age who are known to be
Adverse Events pregnant or who are attempting to become pregnant should not be
Rubella vaccines usually are well tolerated. The most frequent complaints vaccinated. Vaccinated women should be counseled about the need to
after vaccination are fever, lymphadenopathy, or rash, which occur in avoid pregnancy for 28 days after receiving vaccination.471 Although
5% to 15% of children 5 to 12 days after receiving vaccination.44 Transient pregnancy is a contraindication to administering rubella vaccination,
peripheral neuritis (i.e., paresthesia and pain in the arms and legs) has the maximal theoretical risk to the fetus is estimated to be 2.6%.545
been observed uncommonly, primarily in older age groups.580 From 1979 until 1989, the CDC registered 321 susceptible women who
Approximately 3% of children have transient joint manifestations, inadvertently had received RA 27/3 rubella vaccine within 3 months
including arthralgia and, less commonly, arthritis 1 to 3 weeks after before or after conception and carried their pregnancies to term. None
being immunized. Although 25% of women report having joint pain of their infants had defects compatible with CRS, although 2% had
after being vaccinated, arthritis with objective clinical findings lasting serologic evidence of intrauterine infection.113 Because rubella virus
less than 10 days occurs in 13% to 15%. Cases of persistent or recurrent has been isolated from the products of conception of women vaccinated
joint symptoms have been reported but are rare events. In 1992, the during pregnancy, continued caution with respect to vaccination during
IOM reviewed the existing data on rubella and adverse joint events and pregnancy is advised. However, the evidence available indicates that
concluded that the evidence available was consistent with a causal rubella vaccination inadvertently given during pregnancy ordinarily
relationship between rubella vaccination and chronic arthritis in women, does not represent a reason to consider interruption of pregnancy.
although data on current vaccine strains are limited.354 The incidence Although vaccine virus may be isolated from the pharynx, vaccinees
of joint manifestations after immunization is lower than that after natural do not transmit rubella to others, except in the case of a vaccinated
infection at the corresponding age. breastfeeding mother. In this situation, the infant may be infected through
Rubella revaccination is well tolerated, even among college-aged breast milk, and a mild rash illness may develop, but serious adverse
and older vaccinees, and it is associated with a much lower incidence effects have not been noted. Infants infected through breastfeeding
of adverse reactions than primary rubella immunization of young respond normally to rubella vaccination at 15 months of age. Breastfeed-
adult populations. Reported rates of joint-related complaints of 4% to ing is not a contraindication to receiving rubella vaccination.
18% after revaccination are lower than those reported after primary Concern about potential transmission of disease from immunized
vaccination.186,590 children to susceptible contacts, including pregnant women, has not
been supported by studies of susceptible household contacts. Susceptible
Indications children whose household contacts are pregnant may be vaccinated.
Live virus rubella vaccine usually is recommended for all children 12 People with a history of thrombocytopenia may experience throm-
months or older. It is given as MMR or MMRV when children are 12 bocytopenia after receipt of MMR vaccine. The decision to vaccinate
to 15 months of age.44,471 A second dose of rubella vaccine administered should depend on the benefits of immunity compared with the risk of
as MMR or MMRV is given at the time they enter school, usually 4 to recurrence or exacerbation of thrombocytopenia after vaccination or
6 years of age, according to recommendations for routine measles during natural infection with measles or rubella.
immunization. The vaccine should be provided to previously unim- Rubella vaccine should not be given during an interval beginning
munized preschool-aged children or older schoolchildren despite a 2 weeks before and extending 3 months after the administration of
CHAPTER 245  Active Immunizing Agents 2583

immune globulin or blood transfusion. Because rubella vaccine usually contained in the vaccine rather than to response to tetanus toxoid.47
is given as MMR or MMRV, and evidence suggests that high doses of Hypersensitivity reactions can occur in adolescents and adults but rarely
immune globulin preparations can inhibit the immune response to are severe. Neurologic reactions occurring after the administration of
measles vaccine for 3 or more months, depending on the dosage, rubella tetanus toxoid are rare events. Reactions include brachial neuritis and
vaccination with MMR or MMRV necessitates deferral for longer periods Guillain-Barré syndrome.
(see Measles Vaccine).44,471,603 Regarding adverse reactions reported after Tdap, vaccination with
Boostrix was associated with a statistically higher rate of moderate to
Tetanus Toxoid severe headache compared with Td, and that with Adacel was associated
The efficacy of active immunization against tetanus was demonstrated with higher rates of mild injection site pain and low-grade fever compared
most dramatically in military personnel during World War II, when tetanus with the Td vaccine. No serious adverse events have been reported for
toxoid virtually eliminated tetanus in injured servicemen. Since the 1940s, Boostrix. Two cases of serious adverse advents (both characterized as
routine immunization of civilians in the United States with tetanus toxoid neuropathic reactions) in adults possibly related to having received
has been successful in almost eliminating tetanus. In most cases, disease Adacel (none reported in adolescents) have been reported, and in both
has been reported in unimmunized or inadequately immunized individu- cases, symptoms resolved completely within several days.90,405
als.114 The primary three-dose series of a tetanus toxoid–containing vaccine
confers protective immunity for 10 years or longer.90,405 Indications
Without a tetanus booster, immunity wanes over time. The potential Preexposure indications.  For primary immunization, doses of tetanus
for occurrence of tetanus is indicated by the significant number of toxoid provided as DTaP should be administered when the child is 2,
adults in the United States who lack protective concentrations of serum 4, and 6 months of age.21,47,90 A fourth dose is given 6 to 12 months
antibody.289 Between 2001 and 2008, 233 cases of tetanus were reported after the third dose (i.e., at 12 to 18 months of age) to maintain adequate
in the United States with a case-fatality rate of 13%. In 22 of these serum antibody concentrations for the ensuing preschool years. For
cases, the children were between the ages of 5 and 19 years, and 16 had children younger than 7 years not immunized in infancy, an initial dose
received one or no tetanus immunizations. There were no deaths in of DTaP is given with subsequent doses 2, 4, and 10 to 16 months later,
this age group.168 followed by a single booster dose at age 4 to 6 years, just before school
Neonatal tetanus also has been almost eliminated in the United entry.
States. However, it is a leading cause of morbidity in neonates in In 2005, the FDA licensed two Tdap vaccines for routine use in
developing countries. As a result, global elimination of neonatal tetanus adolescents and adults. Adacel (Sanofi Pasteur) is approved for people
remains a goal of the WHO. Because the spores of tetanus are distributed 11 to 64 years of age, and Boostrix (GlaxoSmithKline Biologicals) is
widely in the environment, the risk of infection among the unimmunized approved for children and adolescents 10 years or older. These vaccines
is constant. This is a fact not necessarily appreciated by parents who are approved for one-time use only in individuals who have previously
refuse immunization in the belief that widespread immunization in completed their primary vaccination series with DTP or DTaP as a
the general population will confer some protection on the unimmunized, substitute for their next Td booster.171 The preferred age for Tdap
as was demonstrated by two cases of tetanus in children in Oklahoma vaccination is 11 to 12 years. Catch-up vaccination with Tdap is encour-
in 2012.246,375 aged for adolescents 13 to 18 years old who completed their primary
series with DTP or DTaP and have not received a Td or Tdap booster.
Preparations Adolescents 11 through 18 years of age who received Td but not
Tetanus toxoid is prepared by formaldehyde treatment of Clostridium Tdap are encouraged to receive a single dose of Tdap to provide protec-
tetani toxin.90,405 It has been prepared in fluid and aluminum salt-adsorbed tion against pertussis if they have completed the recommended childhood
preparations, but in the United States, only the latter is available. Fluid DTP/DTaP immunization series. Tdap should be administered regardless
toxoid preparations result in a significantly shorter duration of immunity of the interval since the last tetanus or diphtheria toxoid–containing
than that induced by aluminum-adsorbed antigens; therefore, adsorbed vaccine. Although longer intervals between Td and Tdap may decrease
antigens are recommended. the occurrence of local reactions, the benefits of protection against
Tetanus toxoid is available in combination with diphtheria toxoid pertussis outweigh the potential risk of adverse events.47,171 Every 10
and acellular pertussis vaccine (DTaP) for routine administration to years thereafter, tetanus booster should be provided by Td.
infants and children younger than 7 years and for adolescents and adults Adolescents between the ages of 11 and 18 years who have never
11 years or older as a single booster (Tdap). For all people for whom been vaccinated against tetanus, diphtheria, or pertussis should receive
pertussis vaccine is contraindicated, tetanus toxoid is combined with Tdap initially, followed by Td for the subsequent two doses 4 or more
diphtheria toxoid as DT or Td (see Diphtheria Toxoid). weeks and then 6 to 12 months later. For children between the ages of
The first two Tdap vaccines approved for use in adolescents and 7 and 10 years who have not been vaccinated previously, Tdap vaccine
adults in the United States—Boostrix (GlaxoSmithKline Biologicals) should substitute an initial dose of Td in the catch-up series, followed
and Adacel (Sanofi Pasteur)—were licensed in 2005. Preparations of by two subsequent doses of Td vaccine at 4 or more weeks, and 6 to
DT, Td, and Tdap are identical in the amounts of tetanus toxoid they 12 months after the previous dose.47,171 Interruption of the recommended
contain, but they differ in the quantity of diphtheria toxoid. The dose schedule or delay in administering subsequent doses during primary
of Tdap is the same as that of DT or Td (0.5 mL) and is administered immunization does not reduce immunity.
intramuscularly. Antepartum indications.  In areas of the world where the risk of acquiring
neonatal tetanus is significant, previously unimmunized, pregnant women
Immunogenicity and Efficacy should receive two antepartum doses of a tetanus toxoid–containing
Adequate primary immunization provides sufficient protective titers vaccine spaced at least 4 weeks apart. The second dose should be given
of antitoxin for at least 10 years and ensures prompt, anamnestic at least 2 weeks before delivery. The woman should complete the three-
responses to subsequent booster injections. Immune response to dose series in 6 to 12 months.318,405 Tdap should be substituted for the
tetanus toxoid was measured after administration of each adult Tdap first Td dose if Tdap has not been administered previously. In the
vaccine and compared with that elaborated after Td vaccine. Seroprotec- United States, immunization with Tdap is recommended during each
tive anti-tetanus antibody concentrations, defined as a titer 0.1 IU/mL pregnancy, preferably in the early third trimester.171,175 If vaccination is
or greater, and booster response rates to tetanus were determined to not provided before delivery or during pregnancy, women who have
be noninferior after vaccination with Tdap compared with Td not received Tdap vaccine previously should receive a single dose of
vaccination.90,405 Tdap in the immediate postpartum period, preferably before being
discharged from their hospital or birthing center.
Adverse Events Postexposure wound management.  The potential need for immuno-
Local reactions of pain, swelling, and induration can occur, but these prophylaxis is an integral aspect of wound management at the time
reactions in children usually are attributable to the pertussis component of trauma or injury. The recommended use of tetanus toxoid in
2584 PART V  Prevention of Infectious Diseases

Approximately 90% of cases were children, with the highest incidence


TABLE 245.17  Recommended Tetanus for children 1 to 6 years old.
Prophylaxis in Wound Management After introduction and routine use of varicella vaccine in 1995, the
CLEAN, MINOR ALL OTHER incidence of varicella fell by 90%, deaths from varicella declined by
WOUNDS WOUNDSa 66%, and rates of hospitalization for varicella decreased by 80%.322,504,715
However, a high frequency of breakthrough varicella (i.e., chickenpox
History of Td or Td or occurring in a previously vaccinated person) in immunized children
Tetanus Toxoid Tdapb TIG Tdapb TIG and continuing outbreaks of varicella in schools and in childcare centers
<3 doses or unknown Yes No Yes Yes occurred, despite high rates of vaccination.292 Studies showed that over
≥3 doses Noc No Nod No time the vaccine’s effectiveness was less than 90%,592 and in one study
a
of healthy children, the rate of seroconversion after one dose of the
Including wounds contaminated with dirt, feces, soil, and saliva; puncture wounds;
vaccine was only 76%.477 In June 2006, the CDC and AAP recommended
avulsions; and wounds resulting from missiles, crushing, burns, and frostbite.
b
For children <7 y; diphtheria–tetanus–acellular pertussis (DTaP) or diphtheria-tetanus (DT)
a routine two-dose varicella vaccination policy for children (i.e., first
(depending on vaccine status of patient) is preferred to tetanus toxoid (TT) alone. Tdap is dose at 12 to 15 months and second dose at 4 to 6 years) and catch-up
preferred over Td for underimmunized children ≥7 y who have not received Tdap vaccinations for children, adolescents, and adults who previously had
previously.47 Td is preferred to TT for adolescents who received Tdap previously or when received only one dose.23,451
Tdap is not available. Since recommendation of a routine second dose of vaccine in 2006,
c
Yes if ≥10 y since last tetanus-containing vaccine dose. the incidence of varicella has declined further among children. Data
d
Yes if ≥5 y since last tetanus-containing vaccine dose. More frequent boosters are not show that administration of two doses of varicella vaccine is associated
needed and can accentuate side effects. with higher antibody titers, presumably offering better protection from
Td, Tetanus-diphtheria; Tdap, tetanus–diphtheria–acellular pertussis; TIG, tetanus
varicella.415 Results from a controlled study of the effectiveness of two
immune globulin.
doses of varicella vaccine indicated that administration of two doses
was highly effective in preventing varicella in the first 2.5 years after
implementation of the two-dose schedule to prevent disease.595 Odds
addition to tetanus immune globulin at the time of injury is given in of developing varicella were 95% lower for children who received two
Table 245.17. To ensure adequate immunity, children and adults receiving doses compared with one dose of varicella vaccine.
tetanus toxoid for wound management should be given age-appropriate
preparations of vaccines containing diphtheria and pertussis, unless Preparations
contraindicated, and tetanus toxoid. Tdap is preferred over Td if the Three varicella-containing vaccines are approved for use in the United
person has not received Tdap previously.47,171 Specific recommendations States: varicella vaccine (Varivax), combination MMRV vaccine
depend on the individual’s immunization status, the nature of the wound, (ProQuad), and herpes zoster vaccine (Zostavax). Varicella vaccine was
and the duration of time between when the injury occurred and evalu- licensed in the United States in 1995. It is a preparation of the Oka
ation and treatment were undertaken. strain of varicella-zoster virus (VZV) obtained from the vesicle fluid
After prophylaxis is provided, primary immunization should be of a healthy child with varicella that has been attenuated by serial
completed subsequently in those lacking the recommended number of propagation in human embryo lung fibroblasts, guinea pig embryonic
doses. This conservative approach to the frequent administration of cells, and human diploid cell cultures. The vaccine contains trace amounts
booster doses of tetanus toxoid in wound management for previously of neomycin, fetal bovine serum, sucrose, residual components of human
immunized people is supported by the prolonged immunity from tetanus diploid (MRC-5) cells, and gelatin. The vaccine does not contain
vaccination and the increased incidence of hypersensitivity reactions preservatives.
associated with receipt of frequent booster injections.529 Patients con- Varicella vaccine is lyophilized and stored frozen at −15°C or colder
valescing from tetanus infection should complete active immunization until reconstituted. Any freezer that reliably maintains an average
because infection often does not confer immunity. temperature of −15°C and has a separate sealed freezer door is acceptable
for storing vaccine. The vaccine also may be stored at refrigerator
Precautions and Contraindications temperature (2°C–8°C) for as long as 72 hours before reconstitution.
A history of having an immediate, severe hypersensitivity reaction to Vaccine stored at 2°C to 8°C that is not used within 72 hours should
tetanus toxoid–containing preparations that is severe or anaphylactic be discarded. Reconstituted vaccine should be stored at room temperature
in type is a contraindication to receiving further vaccination.21,47,90,405 and discarded if it is not used within 30 minutes.
People who experience Arthus-type hypersensitivity reactions after In September 2005, a combined live attenuated MMRV vaccine was
receiving tetanus toxoid or temperature greater than 39.4°C (103°F) licensed for use in children 12 months through 12 years of age.148 The
after a previous dose of a tetanus toxoid–containing preparation usually attenuated measles, mumps, and rubella vaccine viruses in MMRV are
have high serum tetanus antitoxin concentrations and should not be identical and of equal titer to those in the MMR vaccine. The titer of
given doses of Td more frequently than every 10 years, even if they Oka/Merck VZV is higher in MMRV vaccine than in single-antigen
have a tetanus-prone wound.47 If an anaphylactic reaction to a previous varicella vaccine. Each 0.5-mL dose contains a small quantity of
dose of tetanus toxoid is suspected, intradermal skin testing may be hydrolyzed gelatin, human albumin, residual components of MRC-5
helpful in determining whether to discontinue tetanus toxoid cells, neomycin, bovine calf serum, and other buffer and media ingre-
vaccination.367 dients. Unlike single-antigen varicella vaccine, MMRV vaccine cannot
Because tetanus toxoid administration has been associated with be stored at refrigerator temperature. MMRV vaccine must be stored
recurrence of Guillain-Barré syndrome in rare cases,359 the decision to frozen at an average temperature of −15°C or lower for up to 18 months.
give additional doses in people with a history of this syndrome within After reconstitution, the vaccine should be used immediately to minimize
6 weeks after receipt of tetanus toxoid should be based on consideration loss of potency and should be discarded if it is not used within 30
of the benefit of revaccination and the comparative risk of recurrence minutes. The diluent should be stored separately at room temperature
of Guillain-Barré syndrome.128 No physician-diagnosed cases of ana- or in the refrigerator. MMRV vaccine contains no preservative.
phylaxis, Arthus reactions, or Guillain-Barré syndrome in any adolescent In May 2006, the FDA approved herpes zoster vaccine for use in
or adult after either Tdap vaccine have been reported. people 60 years or older. No indications for use of this vaccine in children
exist.
Varicella Vaccine
In the prevaccine era, varicella was endemic in the United States and Immunogenicity and Efficacy
virtually all people acquired varicella by the time they reached adulthood. Varicella vaccine is highly immunogenic in susceptible children.
Varicella infection was responsible for an estimated 4 million cases, Seroconversion has occurred in more than 96% of children 12 months
11,000 hospitalizations, and 100 deaths each year in the United States.133 to 12 years of age after one dose of vaccine.685 Preexisting antibody, if
CHAPTER 245  Active Immunizing Agents 2585

present at 12 months of age, does not appear to interfere with antibody These lesions usually occur within 2 weeks and usually are maculopapular
response. As with other viral vaccines, the antibody response after rather than vesicular.596,693
immunization is lower than that from natural disease. Adolescents and In postlicensure studies, the adverse event most frequently reported
adults have age-related decreases in the ability to develop a primary is a mild vesicular rash that occurs in approximately 5% of vaccinees.133,290
response to varicella virus.291 Seroconversion rates of 78% to 82% after Most of these generalized rashes occur within 3 weeks, and most are
one dose and 99% after two doses have been reported in those older maculopapular. In one study, vesicular rashes that occurred within 2
than 12 years.291,685 weeks of vaccination were more likely to be caused by wild-type varicella,
In studies in the United States and Japan, serum antibodies to varicella whereas rashes that occurred more than 2 weeks after vaccination were
have been detected for as long as 10 to 20 years after immunization in more likely to be caused by the Oka vaccine strain.596
more than 95% of immunized children.59,376 Antibody concentrations Systemic reactions can occur. Fever within 42 days of vaccination
have persisted for at least 1 year in 97% of adults and adolescents who is reported by 15% of children and 10% of adolescents and adults.
were administered two doses of vaccine 4 to 8 weeks apart.291 Cell- Most of these episodes of fever have been attributed to concurrent
mediated immunity to VZV has been detected in 87% of children and illness rather than to the vaccine.
94% of adults 5 years after vaccination.714 Clinical trials of MMRV that compared events that occurred within
MMRV vaccine was licensed on the basis of equivalence of immu- 42 days of receiving MMRV or MMR and varicella vaccine separately
nogenicity of the antigenic components, rather than clinical efficacy. in different anatomic sites found the frequencies of local reactions and
Clinical studies involving healthy children 12 to 23 months of age generalized varicella-like rash similar to those described for varicella
indicated that those who received a single dose of MMRV vaccine vaccine.414 A temperature of 38.9°C (102°F) or higher within 42 days
developed levels of antibody to measles, mumps, rubella, and varicella of vaccination was more common in the MMRV group (22%) than in
similar to those of children who received MMR and varicella vaccines the group that received MMR and varicella vaccine at different sites
concomitantly at separate injection sites.414 (15%). A measles-like rash also occurred more frequently in MMRV
Varicella vaccine has been demonstrated to be highly effective in recipients (3%) than in those receiving separate injections (2%). Fever
preventing varicella in children and in reducing the severity of infection and measles-like rash usually occurred 5 to 12 days after receipt of
if they do become infected. In prelicensure clinical trials, vaccine was vaccination. In a postlicensure study, an increased risk of seizure was
70% to 90% effective in preventing varicella and more than 95% effective seen among children 12 to 23 months of age receiving MMRV vaccine
in preventing severe disease.416,674 Several postlicensure studies have compared with those who received MMR and varicella vaccines given
shown similar results, with vaccine effectiveness ranging from 83% to separately in the 7- to 10-day postvaccination period.394
100% in preventing varicella and 87% to 100% in preventing severe Varicella vaccine is a live virus vaccine and may result in a latent
disease.195,365,654 In follow-up studies, chickenpox developed in 0.2% to infection, similar to that caused by wild-type varicella virus. Consequently,
2.3% of vaccinated children per year after exposure to wild-type varicella zoster caused by the vaccine virus has been reported, mostly among
virus, a rate that does not seem to increase with length of time after vaccinated children. Based on reports to VAERS, the rate of herpes
immunization.661 These vaccine failure cases are mild, with fewer skin zoster after varicella vaccination is 2.6 cases per 100,000 vaccine doses
lesions, lower rates of fever, and faster recovery, and they are less distributed.133 The incidence of herpes zoster after natural varicella
contagious than are moderate to severe cases of varicella.75,593,661,671 These infection in healthy people younger than 20 years is 68 cases per 100,000
infections have been called breakthrough varicella. person-years,320 and for all ages, it is 215 cases per 100,000 person-years.242
Although findings of some studies have suggested otherwise, most However, the latter rates should be compared cautiously because they
investigations have not identified time since vaccination as a risk factor are based on populations monitored for longer periods than the vaccinees
for breakthrough varicella. Some investigations have identified asthma, were monitored.
use of corticosteroids, and vaccination at younger than 15 months as Cases of herpes zoster have been confirmed by PCR to be caused
risk factors for breakthrough varicella.251,286,365,646,656 Breakthrough varicella by vaccine virus and wild-type virus, a finding suggesting that some
infection can result from several factors, including interference of vaccine herpes zoster disease in vaccinees may result from antecedent natural
virus replication by circulating antibody, impotent vaccine resulting varicella infection.133,330 Most cases of herpes zoster that occur after
from storage or handling errors, or inaccurate record keeping. Interference administration of vaccine have been mild and have not been associated
from live viral vaccine administered before varicella vaccine can reduce with complications such as postherpetic neuralgia.
vaccine effectiveness. Transmission of the vaccine virus occurs rarely and most often from
A study of 115,000 children in two health maintenance organizations immunocompromised vaccinees. Of the 15 million doses of varicella
from 1995 to 1999 found that children who received varicella vaccine vaccine distributed, on only three occasions has transmission from
less than 30 days after MMR vaccination had a 2.5-fold increased immunocompetent people been documented by PCR analysis.418,574 All
risk of breakthrough varicella compared with those who received three cases resulted in mild disease without complications. In one case,
varicella vaccine before, simultaneously with, or more than 30 days a child 12 months old transmitted the vaccine virus to his pregnant
after receiving MMR.656 Inactivated vaccines (i.e., DTaP, Hib, IPV, and mother.437,574 The mother elected to terminate the pregnancy, but fetal
hepatitis B) and OPV did not increase the risk of breakthrough varicella tissue tested by PCR was negative for varicella vaccine virus. The other
if they were administered less than 30 days before administration of two documented cases involved transmission from healthy children 1
varicella vaccine. year of age to a healthy sibling 4.5 months of age and to a healthy
For adults and adolescents who have seroconverted, varicella vaccine father.133 Transmission also has occurred from a person with herpes
provides protective efficacy rates of approximately 70% after household zoster caused by vaccine strain virus.94 Transmission has not been
exposure. In the remaining 30%, attenuated disease with fewer skin documented in the absence of a vesicular rash after vaccination. No
lesions and little or no systemic toxicity develops, as in children.291 evidence indicates reversion to virulence of the vaccine strain during
transmission; siblings of leukemic vaccine recipients who acquired
Adverse Events vaccine virus had mild rash in 75% of cases and symptomless seroconver-
Varicella vaccine produces relatively few adverse reactions.596,693 The sion in 25%.55
most common adverse reactions that occur after receipt of varicella
vaccine are local reactions, such as pain, soreness, erythema, and swelling. Indications
Based on information from the manufacturer’s clinical trials of varicella Monovalent varicella vaccine and MMRV have been licensed for use
vaccine, local reactions after receiving the first dose are reported by for healthy children 12 months through 12 years of age.26,23,49,155,450,451
19% of children and by 24% of adolescents and adults.596,693 These local Children in this age group should receive two 0.5-mL doses of varicella
adverse reactions usually are mild and self-limited. vaccine administered subcutaneously, separated by at least 3 months.
A varicella-like rash at the site of injection is reported by 3% of The recommendation for at least a 3-month interval between doses is
children and by 1% of adolescents and adults after receipt of the second based on the design of the studies evaluating two doses in this age
dose. In both circumstances, a median of two lesions has occurred. group; if the second dose inadvertently is administered between 28 days
2586 PART V  Prevention of Infectious Diseases

and 3 months after the first dose, the second dose does not need to be Precautions and Contraindications
repeated. Varicella-containing vaccines are contraindicated for pregnancy, severe
All healthy children routinely should receive the first dose of varicella- febrile illness, known history of anaphylactic reaction to vaccine
containing vaccine at 12 through 15 months of age. The second dose components, and immunodeficiency states (i.e., malignancy, primary
of vaccine is recommended routinely when children are 4 through 6 immunodeficiency disease, immunosuppressive or corticosteroid therapy,
years of age (i.e., before a child enters kindergarten or first grade) but and radiation therapy).26,23,49,155,450,451
can be administered at an earlier age. Because of the minimal potential Varicella vaccine should not be administered to people who have
for increased febrile seizures after the first dose of MMRV vaccine in had an anaphylactic-type reaction to any component of the vaccine,
children 12 through 15 months of age, the AAP and ACIP recommend including gelatin and neomycin. Most people with allergy to neomycin
a choice of MMR plus monovalent varicella vaccine or MMRV for have resulting contact dermatitis, a reaction that is not a contraindication
toddlers receiving their first immunization of this kind.26,450 Parents to immunization. Monovalent varicella vaccine does not contain preserva-
should be counseled about the rare possibility of their child developing tives or egg protein, and although the measles and mumps vaccines
a febrile seizure 1 to 2 weeks after immunization with MMRV for the included in MMRV vaccine are produced in chick embryo culture, the
first immunizing dose. For the second dose at 4 through 6 years of age, amounts of egg cross-reacting proteins are not significant. Children
MMRV usually is preferred over MMR plus monovalent varicella to with egg allergy routinely can be given MMRV without previous skin
minimize the number of injections. Varicella vaccine should be admin- testing.
istered to all children in this age range unless there is evidence of Varicella vaccine should not be administered routinely to children
immunity to varicella or a contraindication to administration of the who have congenital or acquired T-lymphocyte immunodeficiency,
vaccine. including those with leukemia, lymphoma, and other malignant neo-
A catch-up second dose of varicella vaccine should be offered to all plasms affecting the bone marrow or lymphatic systems, and children
children 7 years and older who have received only one dose. A routine receiving long-term immunosuppressive therapy. An exception includes
health maintenance visit at 11 through 12 years of age is recommended certain children infected with HIV (discussed later). Children with
for all adolescents to evaluate immunization status and administer impaired humoral immunity can be immunized.
necessary vaccines, including the varicella vaccine. Immunodeficiency should be excluded before immunization of
Individuals 13 years or older without evidence of immunity should children with a family history of hereditary immunodeficiency. An
receive two 0.5-mL doses of varicella vaccine separated by at least 28 immunodeficient or HIV-seropositive household family member does
days. The recommendation for at least a 28-day interval between doses not contraindicate vaccine use in other family members.
is based on the design of the studies evaluating two doses in this age In people with possible altered immunity, immunization against
group. For people who previously received only one dose of varicella chickenpox should use only monovalent varicella vaccine. The Oka vaccine
vaccine, a second dose is necessary. Only monovalent varicella vaccine strain remains susceptible to acyclovir. If a high-risk patient develops
is licensed for use in this age group. vaccine-related varicella, acyclovir should be used as treatment.
Women should be assessed prenatally for evidence of varicella Before routine immunization of healthy children against varicella
immunity. On completion or termination of their pregnancies, women was instituted in the United States in 1995, many young children with
who do not have evidence of varicella immunity should receive the leukemia were susceptible to chickenpox. Considering the variation in
first dose of varicella vaccine before discharge from the health care intensity of chemotherapy regimens and the decreasing incidence of
facility. The second dose should be administered 4 to 8 weeks later at varicella in the United States, these high-risk children should not be
the postpartum or other health care visit. To ensure administration of immunized routinely. Immunization of susceptible leukemic children
varicella vaccine, standing orders are recommended for health care without evidence of immunity in remission should be undertaken only
settings where completion or termination of pregnancy occurs. with expert guidance and with availability of antiviral therapy in case
Postexposure prophylaxis.  Data from the United States and Japan in complications occur.
a variety of settings indicate that varicella vaccine is 70% to 100% Live virus vaccines usually are withheld for an interval of at least 3
effective in preventing illness or modifying the severity of illness if it months after immunosuppressive cancer chemotherapy has been dis-
is used within 3 days and possibly up to 5 days after exposure.573,672 The continued. However, the interval until immune reconstruction varies
ACIP recommends administration of varicella vaccine to people without with the intensity and type of immunosuppressive therapy, radiation
evidence of immunity 12 months or older, including adults, as soon as therapy, underlying disease, and other factors. It often is not possible
possible within 72 hours and possibly up to 120 hours after varicella to make a definitive recommendation for an interval after cessation of
exposure.451 Varicella vaccine may prevent or modify disease and should immunosuppressive therapy when live virus vaccines can be administered
be considered in these circumstances if there are no contraindications safely and effectively.
to its use. A second dose should be given at the age-appropriate interval Screening for HIV infection is not indicated before routine VZV
after the first dose. Physicians should advise parents and their children immunization. Varicella vaccine should be considered for nonimmune
that the vaccine may not protect against disease in all cases because HIV-infected children with a CD4+ T-lymphocyte percentage of 15%
some children might have been exposed at the same time as the index or greater, especially if they are receiving antiretroviral therapy.482 Eligible
case. However, if exposure to varicella does not cause infection, post- children should receive two doses of monovalent varicella vaccine with
exposure immunization with varicella vaccine will result in protection a 3-month interval between doses and return for evaluation if they
against subsequent exposure. There is no evidence that administration experience a postimmunization varicella-like rash. Hundreds of HIV-
of varicella vaccine during the presymptomatic or prodromal stage of infected children have been safely immunized in the United States, and
illness increases the risk of vaccine-associated adverse events or more the vaccine is tolerated much as it is in healthy immunized children.
severe natural disease. Varicella vaccine has protected these children against varicella and herpes
In 2006, the ACIP approved a revised definition for evidence of zoster by preventing infection with wild-type VZV.
immunity to varicella.451 Revised criteria for evidence of immunity to Varicella vaccine should not be administered to people who are
varicella include any of the following: receiving high doses of systemic corticosteroids (i.e., 2 mg/kg per day
• Documentation of age-appropriate vaccination or more of prednisone or its equivalent or 20 mg/day of prednisone
• Preschool-aged children 12 months or older: one dose or its equivalent) for 14 days or more. The recommended interval
• School-age children, adolescents, and adults: two doses between discontinuation of corticosteroid therapy and immunization
• Laboratory evidence of immunity or laboratory confirmation of with varicella vaccine is at least 1 month. Varicella vaccine may be
disease administered to people receiving inhaled, nasal, and topical corticosteroids.
• Birth in the United States before 1980 The results of one small study indicate that two doses of the varicella
• Diagnosis of varicella or verification of history of varicella disease vaccine in 29 children with nephrotic syndrome between 12 months
by a health care provider and 18 years of age usually were well tolerated and immunogenic,
• History of herpes zoster based on health care provider diagnosis including children receiving low-dose, alternate-day prednisone.
CHAPTER 245  Active Immunizing Agents 2587

Household contacts of immunocompromised people should be Immunization Schedule in the United States, a minimum of 22 separate
immunized if they have no evidence of immunity to decrease the injections are needed to immunize a child from birth to 2 years of
likelihood that wild-type VZV will be introduced in the household. age.210,560 At some visits, the administration of three to five separate
Transmission of vaccine-strain VZV from healthy people has been injections can be indicated.
documented in seven instances, resulting in eight secondary cases. Even Combination vaccines represent one solution to the problem of
in families with immunocompromised people, including people with increased numbers of injections. These vaccines incorporate into a single
HIV infection, no precautions are needed after immunization of healthy product antigens that prevent several diseases. Combinations licensed
children in whom a rash does not develop. Immunized people in whom in recent years in the United States are shown in Table 245.18.
a rash develops should avoid direct contact with immunocompromised Licensed combination vaccines can be used when any components
hosts without evidence of immunity for the duration of the rash. of the combination are indicated and its other components are not
Receipt of antibody-containing blood products (i.e., whole blood, contraindicated and if licensed by the FDA for that dose in the series.408
plasma, or parenteral immune globulin) may interfere with seroconver- Use of combination vaccines can reduce the number of injections patients
sion to varicella vaccine. The length of time that such passively acquired receive and alleviate parental concerns about the number of injections.
antibody persists depends on the concentration and quantity of the Potential advantages of combination vaccines include improved vaccine
blood product received408 (see Table 245.11). coverage rates, timely vaccination coverage for children who need
Although no direct evidence demonstrates that varicella vaccine is catch-up vaccination, reduced shipping and stocking costs, reduced
harmful to a pregnant woman or her fetus, the vaccine should not be costs for extra health care visits necessitated by deferral of vaccination,
administered to women known to be pregnant or considering becoming and facilitation of adding new vaccines into vaccination programs.
pregnant within the month because of the theoretical risk of fetal Potential disadvantages of combination vaccines include adverse
infection associated with a live virus vaccine. Vaccinated women should events that may occur more frequently after administration of a combina-
avoid conception for 1 month after receiving vaccination. tion vaccine compared with administration of separate antigens at the
The manufacturer established a Varicella Vaccination in Pregnancy same visit, such as those that occur with the MMRV vaccine and
Registry to monitor the maternal-fetal outcomes of pregnant women combination DTaP-hepatitis B-IPV vaccine450,637; confusion and uncer-
inadvertently given varicella vaccine. The registry was closed in October tainty about selection of vaccine combinations and schedules for
2013 because the low rate of exposure of varicella-susceptible women subsequent doses, especially when vaccinations are given by multiple
of childbearing age to VZV-containing vaccines, in addition to the rarity providers who might be using different products; reduced immunogenic-
of the outcome, contributed to the low feasibility of the registry providing ity of one or more components238; extra doses of certain antigens in
more robust data on the risk of congenital varicella syndrome within the fixed product (e.g., a provider who uses DTaP-hepatitis B-IPV vaccine
a reasonable timeframe.453 A summary of 18 years of registry data found will give an extra dose of hepatitis B component); and a shorter shelf-life
no cases of congenital varicella syndrome and no increased prevalence than the individual component vaccines. The economic impact of the
for other birth defects after exposure to VZV-containing vaccines during use of combination vaccines is unclear because combination products
pregnancy. However, the number of exposures was insufficient to exclude have the potential for increased or decreased costs compared with
a very low risk of congenital varicella syndrome in varicella-susceptible single-antigen component vaccines.
women exposed during the high-risk period.689 The minimum age for administration of a combination vaccine is
A study of nursing mothers and their infants showed no evidence the oldest minimum age for any of the individual components; the
of excretion of vaccine strain in human milk or of transmission to minimum interval between doses is equal to the greatest minimum
infants who are breastfeeding. Varicella vaccine should be administered interval of any of the individual components. When patients have received
to nursing mothers who lack evidence of immunity. the recommended immunizations for some of the components in a
Reye syndrome has occurred in children infected with varicella who combination vaccine, administering the extra antigens in the combination
receive salicylates. Whether varicella vaccine may induce Reye syndrome vaccine is permissible if they are not contraindicated and doing so will
is not known, but the vaccine manufacturer recommends that salicylates reduce the number of injections required. However, excessive doses of
not be given within at least 6 weeks after administration of varicella toxoid vaccines (i.e., diphtheria and tetanus) can result in extensive
vaccine. local reactions.
Only combination vaccines licensed by the FDA should be used.
Diseases for Which Combination Vaccines Are Available Separate vaccines should not be combined into the same syringe for
Many new and improved vaccines have been introduced over the past administration together unless mixing is indicated for the patient’s age
20 years. Incorporation of these vaccines into already complex childhood and is explicitly specified on the FDA-approved product label inserts.
immunization schedules has posed a challenge. In the 2016 Recommended Only one combination vaccine (i.e., DTaP-IPV/Hib vaccine [Pentacel])

TABLE 245.18  Combination Vaccines Licensed in the United Statesa


Vaccineb Trade Name (year licensed) Age Range Routinely Recommended Ages
DTaP-HepB-IPV Pediarix (2002) 6 wk–6 y Three-dose series at 2, 4 and 6 mo
DTaP-IPV/Hib Pentacel (2008) 6 wk–4 y Four-dose series at 2, 4, 6, and 15–18 mo
DTaP-IPV Kinrix (2008) 4–6 y Fifth dose of DTaP and fourth dose of IPV
DTaP-IPV Quadracel (2015) 4–6 y Fifth dose of DTaP and fourth or fifth dose of IPV
HepA-HepB Twinrix (2001) ≥18 y Three doses on a schedule of 0, 1, and 6 mo
Hib-MenCY MenHibrix (2012) 2 –23 mo Four-dose series at 2, 4, 6, and 15–18 mo
MMRV ProQuad (2005) 12 mo–12 y Two doses, the first at 12–15 mo, the second at 4–6 y
a
Excludes measles-mumps-rubella (MMR), DTaP, Tdap, Td, and IPV vaccines, for which individual components are not available. DTaP/Hib (TriHIBit) and Hib/HepB (Comvax) are no longer
manufactured.
b
A hyphen (-) between vaccine products indicates that products are supplied in their final form by the manufacturer and do not require mixing or reconstitution by the user. A slash (/)
indicates that the products must be mixed or reconstituted by the user.
DTaP, Diphtheria–tetanus–acellular pertussis; HepA, hepatitis A; HepB, hepatitis B; Hib, Haemophilus influenzae type b; IPV, inactivated poliovirus; MenCY, meningococcal group CY; MMRV,
measles-mumps-rubella-varicella.
Modified from American Academy of Pediatrics. Combination vaccines. In: Kimberlin DW, Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious
Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:36–7.
2588 PART V  Prevention of Infectious Diseases

contains separate antigen components for which FDA approves mixing Immunogenicity and Efficacy
by the user. The safety, immunogenicity, and efficacy of unlicensed BCG is used primarily in young infants in an attempt to prevent dis-
combinations are unknown. seminated and other life-threatening manifestations of M. tuberculosis
Licensure of a combination vaccine by the FDA is based on studies disease. However, BCG does not prevent infection with M. tuberculosis.
demonstrating that the product’s immunogenicity (or efficacy) and The efficacy of different BCG vaccines seems to vary broadly. Two
safety are comparable or equivalent to monovalent or combination meta-analyses of published clinical trials and case-control studies
products licensed previously. FDA licensure also indicates that a concerning the efficacy of BCG vaccines concluded that BCG has relatively
combination vaccine may be used interchangeably with monovalent high protective efficacy (approximately 80%) against meningeal and
formulations and other combination products with similar component miliary tuberculosis in children.207,562 The protective efficacy against
antigens produced by the same manufacturer to continue the vaccination pulmonary tuberculosis, however, differed significantly among the studies,
series. For example, DTaP, DTaP/Hib, and future DTaP vaccines precluding a specific conclusion. Protection afforded by BCG in one
that contain similar acellular pertussis antigens from the same manu- meta-analysis was estimated to be 50%.207
facturer may be used interchangeably if licensed and indicated for the
patient’s age.152 Adverse Events
Licensure of a vaccine by the FDA does not necessarily indicate that Four billion doses of BCG have been administered with a proven safety
the vaccine is interchangeable with products from other manufacturers. record.383 BCG vaccination usually results in scarring at the site of
Data are readily available for diseases with known serologic correlates injection. However, the vaccine has been associated uncommonly (1–2%
of protective immunity. For diseases without surrogate laboratory of vaccinations) with local adverse reactions such as subcutaneous
markers, prelicensure field vaccine efficacy (phase III) trials or postli- abscess and lymphadenopathy, which are not usually serious.
censure surveillance usually are required to determine protection.478 Osteitis affecting the epiphyses of long bones is a rare complication
The ACIP prefers that doses of vaccine in a series come from the same that develops in 1 of 1 million vaccinees and may occur as long as
manufacturer; however, if this is not possible or if the manufacturer several years after receiving BCG immunization. The rate may be higher
of doses of vaccine given previously is unknown, providers should among neonates. Disseminated fatal disease occurs rarely (0.1–1 case/1
administer the vaccine that they have available. million vaccinees), primarily in people with severely impaired immune
systems.127,409,438,621
Antituberculosis therapy, except for pyrazinamide, is recommended
VACCINES WITH SELECTIVE INDICATIONS FOR to treat osteitis and disseminated disease caused by BCG. Some experts
CHILDREN AND ADOLESCENTS also recommend treatment of chronic suppurative lymphadenitis caused
by BCG. People with complications caused by BCG should be referred
Bacille Calmette-Guérin Vaccine to a tuberculosis expert for management.
Effective control of tuberculosis in the United States has been achieved
by the early identification and treatment of cases, followed by surveillance Indications
of household and other close contacts and institution of appropriate In the United States, administration of BCG should be considered only
preventive measures for those at high risk for disease development. In in limited and select circumstances, such as unavoidable risk of exposure
the United States, the mainstay of preventive therapy is isoniazid to M. tuberculosis and failure or unfeasibility of other methods of control
chemoprophylaxis, which is used in asymptomatically infected people of tuberculosis. The ACIP and AAP have published recommendations
to prevent the progression of infection to disease. In selected instances, for the use of BCG to control tuberculosis in children.33,169 Healthy
however, the potential for acquiring disease, poor compliance by contacts infants from birth to 2 months of age may be given BCG without
instructed to take chemoprophylaxis, or failure of chemoprophylaxis tuberculin skin testing; thereafter, BCG is given only to children with
may justify the use of immunoprophylaxis.127 a negative tuberculin skin test result. BCG immunization should be
Bacille Calmette-Guérin (BCG) is not usually recommended for considered for infants and children who are not infected with HIV in
immunoprophylaxis in the United States because of the low risk of the following circumstances:
infection with Mycobacterium tuberculosis, the unpredictable effectiveness • The child is exposed continually to a person or people with contagious
of the vaccine against adult pulmonary tuberculosis, and the vaccine’s pulmonary tuberculosis resistant to isoniazid and rifampin, and the
potential interference with tuberculin skin test reactivity. Elsewhere in child cannot be removed from this exposure.
the world, BCG vaccine is used in more than 100 countries and is recom- • The child is exposed continually to a person or people with untreated
mended routinely at birth by the WHO696 (http://www.who.int/ or ineffectively treated contagious pulmonary tuberculosis, and the
immunization/policy/Immunization_routine_table2.pdf?ua=1). Of child cannot be removed from the exposure or given antituberculosis
primary concern to pediatricians is the risk to an infant born to a therapy.
mother with tuberculosis or living within a household with other Careful assessment of the potential risks and benefits of BCG vaccine
identified individuals with tuberculosis.48 and consultation with personnel in local area control programs for
tuberculosis are strongly recommended before the use of BCG. When
Preparations BCG vaccine is given, care should be taken to observe the precautions
BCG is the only approved vaccine against tuberculosis. BCG is a live and directions for administration on the product label. Other childhood
attenuated strain derived from Mycobacterium bovis. All currently vaccines can be administered concurrently with BCG.
available BCG vaccines are derived from the original strain at the Pasteur
Institute in Paris, but they have been propagated by different methods Skin Test Reactivity
in many laboratories and therefore vary in their immunogenic and Recipients of BCG should have repeat tuberculin skin tests 2 to 3 months
reactogenic properties. BCG (Merck) is the only vaccine licensed in the after immunization to establish that tuberculin cellular reactivity has
United States. Comparative evaluations of this preparation and other developed. Failure to react dictates the need for repeat BCG vaccination
BCG vaccines have not been performed. followed by repeat tuberculin testing.127 The tuberculin reaction to the
BCG is administered only percutaneously using a sterile multiple- BCG vaccine available in the United States usually results in 7 to 15 mm
puncture device. The standard dose of BCG vaccine is 0.2 to 0.3 mL of induration after vaccination and diminishes gradually during sub-
of reconstituted lyophilized BCG. At least 10 new potential vaccines to sequent years. Without revaccination or repeated exposure to M.
prevent tuberculosis are in clinical development.274 Nevertheless, BCG tuberculosis, reactivity usually disappears within 10 years.127 The size of
will remain in use in the foreseeable future and may continue to be the area of induration may be correlated with the number of doses of
used as a prime vaccine in a prime-boost immunization schedule in BCG.357 However, tuberculin skin test sensitivity does not correlate with
conjunction with new tuberculosis vaccines.383 BCG preparations instilled BCG efficacy.337 In BCG recipients, differentiating between a tuberculin
in the treatment of bladder cancer are not intended to be used as reaction representing acquired tuberculous infection and persistent
vaccines.127 postvaccination reactivity is difficult.
CHAPTER 245  Active Immunizing Agents 2589

Because the degree and duration of protection against tuberculous volunteers, vaccine efficacy was found to be 90% at 10 days and 80%
disease afforded by BCG are uncertain, a positive tuberculin reaction 3 months after a single vaccine dose.184 Although the CVD 103-HrG
may be indicative of disease. BCG recipients with a positive tuberculin vaccine provides protective vibriocidal antibodies in 90% of vaccinees,
skin test result should be carefully evaluated with risk assessment, it is specific to V. cholerae serogroup 01 and does not confer protection
radiography, and other diagnostic tests, including an interferon-γ release to O139 or other non-O1 serogroups.184 CVD 103-HrG vaccines have
assay (IGRA), which does not produce cross-reactions after sensitization the benefit of being efficacious with a onetime dose instead of the two
by BCG.467 spaced doses required of the killed, whole cell vaccines.
WC/rBS vaccine confers protection specific to V. cholerae serogroup
Precautions and Contraindications O1.345 Immunization does not protect against V. cholerae serogroup
BCG vaccine should not be administered to individuals with burns, skin O139 or other species of Vibrio. Field trials in Bangladesh, Peru, and
infections, and certain primary or secondary immunodeficiencies, Sweden have shown that the WC/rBS vaccine confers 85% to 90%
including HIV infection, because of the risk of disseminated BCG disease. protection for 6 months in all age groups after administration of two
The use of BCG vaccine also is contraindicated in people receiving doses 1 week apart. In Bangladesh, protection declined rapidly after 6
immunosuppressive medications, including high-dose corticosteroids. months in young children, but it was still approximately 60% effective
In the United States, where the risk of acquiring tuberculosis is low, in older children and adults after 2 years. Because of the limited duration
BCG vaccine should not be administered to children with known or of protection, the WC/rBS vaccine is not recommended to be used in
suspected asymptomatic HIV infection.127,510 However, in countries children younger than 2 years.
where the risk of contracting tuberculosis is high and HIV surveillance The modified killed, whole cell–only vaccine had been studied in
of pregnant women and infants is limited, the WHO has recommended several trials and found to be safe and highly effective, with seroconversion
that asymptomatic HIV-infected children receive BCG vaccine at birth rates of vibriocidal antibodies ranging from 53% to 91%.54,572,599,628 Limited
or shortly thereafter.696,695 However, the WHO recommends that BCG data on the safety and immunogenicity of the modified whole-cell–based
should not be administered to infants known to be infected with HIV vaccine in children at least 1 year of age appear promising.572
even in resource-poor regions because the risk of disseminated BCG
in these infants is significantly high, approaching 1%.698,699 Although Adverse Events
no harmful effects of BCG vaccine on the fetus have been documented, In clinical trials of the newly licensed CVD 103-HgR vaccine, there was
women should avoid receiving vaccination during pregnancy. no difference between placebo and vaccine recipients with respect to
diarrhea, asthenia, headache, abdominal pain, anorexia, nausea and
Cholera Vaccine vomiting, or fever.184
The global burden of cholera is difficult to estimate due to underreport- Millions of doses of WC/rBS vaccine have been supplied worldwide.345
ing, but experts place the annual number of cases at 2.9 million with According to manufacturer information from clinical trials and post-
95,000 deaths.8 In 2009, 45 countries reported 221,226 cholera cases marketing surveillance, mild gastrointestinal symptoms (e.g., abdominal
with 4946 deaths to the WHO, with 98% of cases and 99% of deaths pain, cramping, diarrhea, nausea) are most commonly reported, occurring
reported from Africa.566 In, 2010, an outbreak of cholera occurred in at a frequency of 0.1% to 1%. Serious adverse events, including a flulike
Haiti with more than one-half million cases and more than 7000 syndrome, rash, arthralgia, and paresthesias, are rare, occurring in less
confirmed deaths. It was the first epidemic in the Western Hemisphere than one of 10,000 doses distributed. In several RCTs, the modified,
since the 1991 epidemic in Peru.6 After it emerged in Haiti, cholera killed, whole cell–only vaccine also demonstrated a low frequency of
spread to other countries, including the Dominican Republic and Cuba. mild symptoms and no major adverse events.54,572,628
From 2001 through 2013, 123 confirmed cases of cholera in the
United States were acquired abroad; of these, 63 were associated with Indications
the epidemic in Hispaniola.436 Most cases of disease have occurred in In June 2016, the ACIP voted to recommend cholera vaccine (CVD
travelers to cholera-affected areas or people who have eaten contaminated 103-HgR, Vaxchora) for adult (18–64 years old) travelers to an area of
food brought or imported from these areas.122 Although cholera remains active toxigenic V. cholerae O1 transmission. Special consideration should
a significant public health concern in African, Asian, and South American be given for those at higher than average risk, such as health care profes-
countries, even in these countries the risk to US travelers is low. People sionals in endemic areas, aid workers in refugee camps, and those traveling
following the usual tourist itinerary that use standard accommodations to remote areas where cholera epidemics are occurring and access to
in countries reporting cholera are at very low risk for infection.566 medical care is limited. No country requires proof of cholera immuniza-
tion as a condition for entry, and the WHO recommends against such
Preparations a requirement. Some local authorities, however, may require immuniza-
There are two major types of oral vaccines against cholera: killed whole tion; to determine local requirements, travelers should consult the
cell-based and genetically attenuated live vaccines.345 A single cholera embassies of the countries to which they will be traveling.
vaccine CVD 103-HgR (Vaxchora; PaxVax, Redwood City, CA) is available
in the United States. This vaccine is an attenuated live oral genetically Precautions and Contraindications
modified V. cholerae O1 strain (CVD 103-HgR) and was licensed by Oral cholera vaccine should not be administered to people with a
FDA in 2016 for United States travelers 18 to 64 years of age visiting hypersensitivity reaction to any of the vaccine components. Administra-
endemic countries. tion of oral cholera vaccine should be postponed for people with acute
Two other vaccines are prequalified by WHO and are available in gastrointestinal illness or acute febrile illness.
many countries outside the United States: WC/rBS (Dukoral; Crucell,
the Netherlands) and Modified WC-only (Shanchol; Shantha Biotechnics, Japanese Encephalitis Virus Vaccine
Medchal, Hyderabad, India). The WC/rBS vaccine is a whole, killed Japanese encephalitis (JE) virus, the most important cause of epidemic
cell–based vaccine that contains Vibrio cholerae O1 with purified mosquito-borne arboviral encephalitis in Asia, has a wide clinical
recombinant B subunit of cholera toxoid. The modified WC-only vaccine spectrum, ranging from asymptomatic infection to permanent neurologic
is also a whole, killed cell-based vaccine but lacks the B subunit. The sequelae associated with a high rate of disability (i.e., 709,000 disability-
WC-only vaccine was originally manufactured by the government of adjusted life-years), and a high case-fatality rate of 20% to 30% or even
Vietnam. Although inexpensive to produce, it did not elicit antitoxic higher among children younger than 10 years. The annual incidence
immunity. In 2007, the WC-only vaccine was modified with international of JE among 3 billion people at risk in 24 countries in Southeast Asia
efforts to create a low-cost vaccine that is produced in India572,599 and the Western Pacific is 1.8 case per 100,000 people despite the
availability of effective vaccines in many regions.700
Immunogenicity and Efficacy The JE virus is a zoonotic flavivirus that cannot be eliminated because
In a randomized, placebo-controlled, double-blind trial of the CVD its animal reservoirs are widespread. However, human infection could
103-HgR vaccine with an oral cholera challenge in 197 health human be controlled by universal human immunization in endemic areas. The
2590 PART V  Prevention of Infectious Diseases

envelope glycoprotein of the JE virus contains the major epitopes that protective measures (i.e., bed nets and mosquito repellants) to reduce
are targeted by neutralizing antibodies. the risk of mosquito bites. For some travelers who will be in high-risk
Historically, an inactivated mouse brain–derived vaccine (JE-VAX) settings, JE vaccine can further reduce the risk of infection. The CDC
controlled JE virus infection successfully among human populations recommends JE vaccine for travelers who plan to spend a month or
in Japan, Korea, and Taiwan since 1968.517 This vaccine is no longer longer in areas with endemic infection during the JE virus transmission
available in the United States and is being phased out elsewhere because season.30,271 JE vaccine should be considered for shorter-term travelers
of greater reactogenicity, higher cost, and larger number of doses if they plan to travel away from an urban area and have an itinerary
compared with the newer-generation vaccines. or activities that increase the risk of JE virus exposure. Information on
the location of JE virus transmission and detailed information on vaccine
Preparations recommendations and adverse events can be obtained from the CDC
The 15 JE vaccines in use in Asia are classified in four categories: website (https://wwwnc.cdc.gov/travel).
inactivated mouse brain–derived vaccines, inactivated Vero cell–derived Primary immunization with JE-VC should be completed at least 1
vaccines, live attenuated vaccines, and live recombinant (chimeric) week before potential exposure to JE virus. If the primary series was
vaccines. In the United States, the inactivated Vero cell–derived, alum- administered more than 1 year earlier, a booster dose is recommended
adjuvanted vaccine, JE-VC (Ixiaro, Intercell Biomedical, United Kingdom, for individuals older than 16 years before potential reexposure.248 Data
distributed by Novartis Vaccines and Diagnostics in the United States) on the response to a booster dose administered more than 2 years after
is indicated for use in individuals 2 months and older.177 It is derived the primary series and the need for and timing of additional booster
from the attenuated SA 14-14-2 JEV strain. doses are not available. There is no information about the utility of
The primary vaccination series for JE-VC is two intramuscular doses booster doses for children. The sparse available data suggest that the
administered 28 days apart. The dose for children younger than 3 years different JE vaccines are interchangeable without causing adverse reactions
is 0.25 mL, and 0.5 mL is given to children 3 years or older.177 The or compromising immunogenicity.
vaccine does not contain stabilizers, antibiotics, or thimerosal and should
be stored at 2°C to 8°C (35°F–46°F) and protected from light. Precautions and Contraindications
A primary hamster kidney cell—derived, live attenuated vaccine A severe allergic reaction after a previous dose of JE-VC is a contraindica-
(CD.JEVAX) has been used widely in China since 1988 and is used tion to administration of subsequent doses.271 JE-VC contains protamine
widely in Asia now. It is based on the SA 14-14-2 JEV strain. Primary sulfate, a compound known to cause hypersensitivity reactions in some
immunization consists of a single 0.5- mL subcutaneous dose in children people.
8 months or older. It contains gelatin, human serum albumin, and Practitioners should use caution when considering the use of JE
other stabilizers. vaccine in pregnant women. Vaccination with JE vaccines usually should
A live attenuated, recombinant (chimeric) JE virus vaccine (IMOJEV, be deferred because of a theoretical risk to the developing fetus. However,
JE-CV, ChimeriVax-JE) was licensed in Australia in 2010 and is used pregnant women who must travel to an area in which the risk of JE
widely in Asia. The premembrane and envelope coding sequences derived exposure is high should be vaccinated with an inactivated JE vaccine
from the SA 14-14-2 JEV strain were engineered to replace the homolo- if the benefits outweigh the risks of vaccination for the mother and
gous sequences of the live attenuated yellow fever vaccine virus, which developing fetus.
is used as a vector. This vaccine is propagated in Vero cells. Primary Breastfeeding is not a contraindication to vaccination with inactivated
immunization is a single dose of 0.5 mL given subcutaneously at 9 JE vaccine. However, whether JE-VC is excreted in human milk is not
months or older, and a booster dose is indicated for children. known. Because many drugs are excreted in human milk, practitioners
should use caution when considering the use of inactivated JE vaccine
Immunogenicity and Efficacy in breastfeeding women.
The JE-VC vaccine was licensed in the United States on the basis of its No data exist on the use of JE-VC in immunocompromised people
ability to induce JE virus–neutralizing antibodies as a surrogate for or patients receiving immunosuppressive therapies.
protection and safety evaluations in approximately 5000 adults.587 The
accepted immunologic surrogate of protection is a serum neutralizing Rabies Vaccine
antibody titer of at least 1 : 10 as determined by a 50% plaque reduction Rabies is a viral zoonosis transmitted in saliva and other tissue of infected
neutralization assay (PRNT50) result.700 mammals. The virus enters the central nervous system of the host and
Vaccine effectiveness for the inactivated JE-VC vaccine is at least causes an acute, progressive encephalomyelitis that is almost universally
93%, 80% to 99% for the live attenuated JE vaccines, and at least 90% fatal. Postexposure prophylaxis is possible because of the long incubation
for the live recombinant vaccines. Long-term immunogenicity data are period, usually weeks to months, of this infection. The time of incubation
limited. A booster vaccine is recommended 2 years after the primary correlates with the length of the axon along which the virus migrates
series to induce a robust anamnestic response.700 to eventually infect the central nervous system. Children are at particular
Mass immunization campaigns with live attenuated and inactivated risk of exposure to rabies from an infected dog, cat, or other animal
mouse brain–derived JE vaccines significantly reduce JE disease in a because they are less likely to take aversive measures if attacked and
community. No data are available for the inactivated Vero cell–derived are more likely to sustain an injury to the head or neck. Accordingly,
and live recombinant vaccines. children are likely to have shorter incubation periods especially with
an exposure injury on the head or neck.
Adverse Reactions In this century, the number of human deaths in the United States
The inactivated Vero cell–derived vaccines, live attenuated vaccines, attributed to rabies has declined from 100 or more each year to an
and live recombinant vaccines all have acceptable safety profiles. The average of 2 or 3 each year. Two programs have been responsible for
most common local reactions to JE-VC are rash, fever, headache, myalgia, this decline. First, animal control and vaccination programs begun in
and fatigue, but severe reactions occur in 1.6 of 100,000 administered the 1940s and oral rabies vaccination programs in the 2000s have
doses,177,700 as described on the CDC website (http://www.cdc.gov/ eliminated domestic dogs as reservoirs of rabies in the United States.
vaccines/acip/recs/grade/je-child.html). Severe adverse events include Second, effective human rabies vaccines and immunoglobulins have
febrile convulsions, thrombocytopenic purpura, and encephalitis.247 The been developed.
other vaccine preparations are associated with similar reactogenicity Wild animals are the most important source of infection for humans
and tolerability as that of JE-VC. and domestic animals. Insectivorous bats carrying variants of the rabies
virus have been responsible for most human cases of rabies in the
Indications continental United States in recent years. Transmission has occurred
The risk of JE for most travelers to Asia is low but varies on the basis from minor and unrecognized bites from infected bats. Wild carnivores,
of destination, duration, season, and activities. All travelers to countries especially raccoons, skunks, and foxes, are the terrestrial animals most
with endemic JE should be informed of the risks of JE and use personal often infected with rabies. Wildlife rabies occurs throughout the
CHAPTER 245  Active Immunizing Agents 2591

continental United States; only Hawaii remains consistently free of rabies. Indications and Precautions
Domestic animals, including dogs, cats, and ferrets, can be infected, When used as indicated, both types of rabies vaccine are considered
with three times as many reports of rabies annually in cats compared equally safe and effective for preexposure and postexposure prophylaxis
with dogs. as described in the ACIP recommendations.448,570 Usually, an immuniza-
Human exposure to rabies from a domestic animal in the United tion series is initiated and completed with one vaccine product. No
States is a rare occurrence. In most other countries (e.g., Asia, Africa, clinical studies have been conducted that documented a change in efficacy
and Latin America), dogs remain the most important potential source or the frequency of adverse reactions when the series is completed with
of infection. International travelers in areas where canine rabies is a second vaccine product. Ideally, an immunization series should be
endemic are at increased risk for exposure to rabies. completed with the same product unless serious allergic reactions occur.
Rabies was diagnosed in a total of 37 people in the United States Corticosteroids, immunosuppressive conditions, and concurrent
since 2003.485 Seventy percent of them acquired the disease in the United administration of antimalarial agents can interfere with the adequate
States or Puerto Rico. Organ or tissue transplantation was identified development of an active immune response. Postvaccination titers should
as the source of infection for 19% of individuals. Bats were implicated be checked in these circumstances or if a significant interruption in
as the source of infection in 65%, with a bat bite reported in vaccine schedule has occurred, especially in the context of a more
seven cases bat contact without a reported bite in six cases and a rabies significant exposure, and in consultation with local public health
virus associated with bats without a known exposure identified in four authorities.
cases. Thirty percent of human rabies deaths reported to the CDC since For adults, rabies vaccination always should be administered in the
2003 appear to have been related to rabid animals outside the United deltoid area. For children, the anterolateral aspect of the thigh also is
States. acceptable. The gluteal area never should be used for HDCV or PCEC
injections because administration of HDCV in this area results in lower
Preparations neutralizing antibody titers and decreased immunogenicity.272
Two rabies vaccines are available for preexposure and postexposure Postexposure prophylaxis.  The essential components of rabies post-
prophylaxis in the United States: human diploid cell vaccine (HDCV) exposure prophylaxis are wound treatment and, for previously unvac-
and purified chicken embryo cell (PCEC). Both vaccines are licensed cinated people, concurrent administration of human rabies immune
for intramuscular administration. HDCV (Imovax Rabies, Sanofi Pasteur) globulin (HRIG) and vaccine (Table 245.19).448,570 The combination of
is derived from the Pitman-Moore strain grown in human diploid cell active and passive immunization is indicated for the treatment of all
culture. PCEC (RabAvert, Chiron Corporation-Novartis) is prepared bite and all non-bite exposures inflicted by animals suspected or proven
from the fixed rabies virus strain Flury LEP grown in primary culture to be rabid. Recommendations for the management of people with
of chicken fibroblasts. The HDCV and PCEC vaccines each contain the possible exposure to rabies include giving meticulous attention to
WHO-recommended standard of at least 2.5 IU of rabies virus antigen thorough cleansing of the wound with soap and water. The decision
per 1.0-mL intramuscular dose. Although not licensed for use in the to give rabies immunoprophylaxis depends on the circumstances
United States, two intradermal rabies vaccines are recommended by precipitating the exposure, the species and condition of the animal
the WHO and used in countries where the two tissue culture vaccines inflicting the wound, and the prevalence of rabies in local animal
(i.e., HDCV and PCEC) are prohibitively expensive.132 populations. Bite and nonbite exposures, including scratches, abrasions,
open wounds, and mucous membranes contaminated with saliva, are
Immunogenicity and Efficacy considered significant. Because the need for preventive measures is
Viral neutralizing antibodies are produced within 7 to 10 days, and based on these specific circumstances, the local department of health
protective immunity usually persists for 2 years or longer. Although a should be consulted promptly concerning the necessity for initiating
definitive protective titer has not been identified, the WHO and CDC postexposure prophylaxis.
adhere to working guidelines for an acceptable response to immuniza- When possible, the brains of wild animals (i.e., skunks, foxes, coyotes,
tion.118 The CDC specifies complete viral neutralization at a 1 : 5 or raccoons, and bats), stray dogs or cats, or symptomatic animals implicated
greater titer by the rapid fluorescent-focus inhibition test as acceptable; in an exposure should be examined in certified laboratories for evidence
the WHO specifies 0.5 IU/mL or more as acceptable. of rabies. Immunization always should be initiated promptly and
Given the essentially universal fatal prognosis of infection after discontinued only if laboratory results are negative. Individuals exposed
symptoms develop, no randomized, placebo-controlled human trials to healthy dogs or cats that are available for observation do not require
have documented the efficacy of vaccine for preexposure or postexposure immediate prophylactic treatment. Implicated healthy domestic dogs
prophylaxis. However, substantial field experience and direct evidence or cats should be quarantined and observed for at least 10 days. If
from controlled animal studies demonstrate a protective effect. The symptoms develop that suggest rabies, the exposed individual should
paucity of human cases attests to the efficacy of postexposure prophylaxis begin postexposure prophylaxis, and the brain of the animal should be
with the currently recommended vaccine and immune globulin examined. An unknown or unavailable animal must be regarded as
preparations. potentially rabid.
Rabies has not been reported in the United States in any patient Studies conducted in the United States by the CDC have documented
who received the recommended postexposure measures. Cases of human that a regimen of one dose of HRIG and five doses of rabies vaccine
rabies occurring after postexposure prophylaxis have resulted from (1.0 mL for each dose) given on days 0, 3, 7, 14 and 28 was safe and
failure to adhere to established guidelines, such as those of the CDC induced an excellent antibody response in all recipients.448 In March
or the WHO.117,250,272,598,673 2010, the ACIP changed recommendations to one dose of HRIG with
four doses of rabies vaccine (1.0 mL for each dose) given on days 0, 3,
Adverse Events 7, and 14. The number of doses recommended for people with altered
Reactions occurring after administration of HDCV or PCEC vaccine immunocompetence has not changed; they should be administered a
are less serious and less common than those associated with the previously five-dose vaccination regimen (days 0, 3, 7, 14, and 28) along with one
available vaccines.132 Local reactions at the injection site occur in 30% dose of HRIG.570 If anatomically feasible, the full dose of HRIG (20 IU/
to 74% of injections, and mild systemic reactions such as fever, headache, kg) should be infiltrated thoroughly in the area around and into the
nausea, abdominal pain, muscle aches, and dizziness occur in 5%to wounds. Any remaining volume should be injected intramuscularly at
40% of vaccine recipients. In one report, approximately 6% of people a site distant from that of vaccine administration. The dose of HRIG
had an immune complex–like reaction 2 to 21 days after receipt of the should not exceed that recommended and should not be given beyond
booster dose of HDCV.108 This systemic hypersensitivity reaction occurred the seventh day because HRIG can partially suppress active antibody
less frequently in people receiving primary vaccination. The reactions production.
have been associated with the presence of β-propiolactone–altered People previously fully vaccinated do not require HRIG and should
human albumin in HDCV and the development of IgE antibodies to receive only vaccine (two doses 3 days apart) because an amnestic
this allergen.273 response will occur after the administration of a booster regardless of
2592 PART V  Prevention of Infectious Diseases

TABLE 245.19  Rabies Postexposure Prophylaxis for Individuals Not Previously Immunized
Evaluation and Disposition
Animal Type of the Animal Postexposure Prophylaxis Recommendations
Wild
Skunks, raccoons, foxes, and most other Regard as rabid unless animal proven Consider immediate vaccination and if not previously vaccinated, use
carnivores; bats negative by laboratory testsa rabies immune globulin
Domestic
Dogs, cats, and ferrets Healthy and available for 10 days Persons should not begin prophylaxis unless animal develops clinical
observation signs of rabiesb
Rabid or suspected rabid Immediately vaccinate
Escaped (unknown) Consult public health officials
Other
Livestock, small rodents, large rodents Consider individually Consult public health officials
(woodchucks and beavers), lagomorphs Bites of squirrels, hamsters, guinea pigs, gerbils, chipmunks, rats,
(rabbits and hares), and other mammals mice, other small rodents, rabbits, and hares rarely require antirabies
prophylaxis
a
The animal should be euthanized and tested as soon as possible. Holding for observation is not recommended. Discontinue vaccine if immunofluorescence test results of the animal are
negative.
b
During the 10-day observation period, begin postexposure prophylaxis at the first sign of rabies in a dog, cat, or ferret that has bitten someone. If the animal exhibits clinical signs of rabies,
it should be euthanized immediately and tested.
Modified from Manning SE, Rupprecht CE, Fishbein D, et al. Human rabies prevention—United States, 2008: recommendations of the Advisory Committee on Immunization Practices.
MMWR Recomm Rep. 2008;57(RR-3):1–28.

the antibody titer.448 Serum for antibody testing should be obtained of acquiring rabies must be carefully considered before deciding to
from people whose prophylaxis history or immune status is uncertain, discontinue vaccination. Advice and assistance on the management of
and the course of postexposure active and passive immunoprophylaxis serious adverse reactions for people receiving rabies vaccines may be
as described for nonimmune individuals should be initiated immediately. sought from the state health department or the CDC.
If serologic testing demonstrates adequate anti-rabies antibody, post-
exposure prophylaxis can be discontinued. Typhoid Vaccine
Once initiated, rabies prophylaxis should not be interrupted or Typhoid fever, an acute, life-threatening febrile illness caused by the
discontinued because of local or mild systemic adverse reactions to bacterium Salmonella enterica serovar typhi (S. typhi), remains a serious
rabies vaccine. Usually, the reactions can be successfully managed with public health problem throughout the developing world. Although
antiinflammatory and antipyretic agents such as ibuprofen or acet- disease prevalence varies greatly depending on the specific population
aminophen. When a person with a history of serious hypersensitivity studied, current global estimates suggest that 27 million cases of typhoid
to rabies vaccine must be revaccinated, antihistamines can be admin- and 270,000 deaths occur annually.97 In the United States and much of
istered. Epinephrine should be readily available to counteract any the developed world, typhoid fever has virtually disappeared.97 Approxi-
anaphylactic reactions, and the person should be observed carefully mately 400 cases of typhoid fever, mostly among travelers, are reported
immediately after receiving vaccination. to the CDC each year. Most reported cases in the United States are
Preexposure prophylaxis.  Active immunization should be considered acquired during travel to developing countries.374 The primary indication
for high-risk groups (i.e., veterinarians, animal handlers and control for typhoid vaccination in the United States is international travel to
officers, selected laboratory workers, people visiting countries where an endemic area.373 In developing countries without safe water and
rabies is hyperendemic, and people whose pursuits may involve frequent sanitation, mass immunization is a potentially effective strategy to limit
contact with rabid animals, such as spelunkers). People whose risk of the severity and impact of typhoid fever.3
exposure is less but whose access to immediate competent medical care The changing epidemiology of typhoid fever underscores the
is restricted also should be considered for preexposure prophylaxis. importance of vaccination of international travelers and high-risk
The primary series consists of three doses (1.0 mL/dose) administered populations in endemic regions. Studies report that the highest incidence
on days 0, 7, and 21 or 28, given intramuscularly in the deltoid area. rate is found among children younger than 5 years and results in more
The three-dose series provides long-term protective immunity, and severe disease than in earlier periods.604,609 S. typhi is increasingly resistant
routine serologic testing for rabies antibody after primary immunization to ampicillin, chloramphenicol, and cotrimoxazole; even quinolone
is not necessary. For individuals who may be immunosuppressed, resistance is reported in many parts of the world.97,366 Case-fatality rates,
measurement of anti-rabies antibodies should be performed. In the which had decreased from 10% to 1% with appropriate antibiotic therapy,
case of continued or frequent exposure, serum samples should be tested could rise again without safe, effective, and affordable vaccination
every 2 years and a booster dose of rabies vaccine provided when the strategies, especially as rates of resistance increase across the globe.
antibody titer falls below the minimum accepted level.448
Preparations
Precautions and Contraindications The three types of typhoid vaccines are inactivated, whole-cell vaccines;
Because of the potential consequences of inadequately treated rabies live attenuated bacterial vaccines; and subunit vaccines.503 The parenteral,
exposure and because no indication exists that fetal abnormalities have heat-phenol–inactivated, whole-cell vaccine widely used for many years
been associated with rabies vaccination, pregnancy is not considered a was highly immunogenic but also was highly reactogenic and no longer
contraindication to postexposure prophylaxis.448 If the risk of exposure is available in the United States. It is the only vaccine approved for use
to rabies is substantial, preexposure prophylaxis also can be indicated in children as young as 6 months and is still available in several developing
during pregnancy. countries because of its affordability, although the manufacture of this
People who have a history of serious hypersensitivity to rabies vaccine vaccine may not be up to international standards.
should be revaccinated with caution. Although serious systemic, ana- Two vaccines are licensed for use in the United States: an oral live,
phylactic, or neuroparalytic reactions are rare events during and after attenuated vaccine (TY21a, Vivotif Berna) and a purified Vi (virulence)
the administration of rabies vaccines, the reactions pose a serious capsular polysaccharide of S. typhi vaccine (ViCPS) (Typhim Vi, Aventis
dilemma for the patient and the attending physician. A patient’s risk Pasteur) for intramuscular use. Table 245.20 provides information on
CHAPTER 245  Active Immunizing Agents 2593

TABLE 245.20  Dosage and Schedule for Typhoid Fever Vaccination


Vaccination Age (y) Dose/Route of Administration No. of Doses Dosing Schedule Boosting Interval
Oral Live Attenuated TY21a Vaccine
Primary series ≥6 1 capsulea/oral 4 Days 0, 2, 4, 6 NA
Booster ≥6 1 capsulea/oral 4 Days 0, 2, 4, 6 Every 5 y
Vi Capsular Polysaccharide Vaccine
Primary series ≥2 0.50 mL/IM 1 NA NA
Booster ≥2 0.50 mL/IM 1 NA Every 2 y
a
Administer with cool liquid no warmer than 37°C (98.6°F).
NA, Not applicable.
Modified from Jackson BR, Iqbal S, Mahon B, et al. Updated recommendations for the use of typhoid vaccine—Advisory Committee on Immunization Practices, United States, 2015. MMWR
Morb Mortal Wkly Rep. 2015;64:305–8.

vaccine dosage and administration. The time required for primary is administered parenterally, mucosal immunity does not develop. The
vaccination is different for the two vaccines, as are the lower age limits efficacy of ViCPS vaccine in clinical trials was 72% at 17 months2 and
for use in children. 64% at 21 months397 after administration of a single dose. A cluster-
Ty21a is an oral, live attenuated vaccine consisting of a stable mutant, randomized trial in India enrolling 37,673 children 2 years or older
Ty21a, developed by chemical mutagenesis of a pathogenic S. typhi demonstrated 61% effectiveness.629 Polysaccharide vaccines do not
strain. This vaccine was licensed in the United States in 1989 for use stimulate T cells and cannot establish immunologic memory. Additional
in adults and children 6 years or older and is available as an enteric-coated doses of ViCPS therefore do not elicit a booster effect. Revaccination
capsule (i.e., must be swallowed whole). The four-dose primary vaccine with a single dose is recommended every 2 years for people who remain
series in the United States and Canada (three-dose series in other parts at risk.
of the world) is administered as one capsule every other day, taken 1 Conjugate Vi vaccines trials have used various carrier proteins, and
hour before a meal, for a total of four capsules. The capsules should one study of more than 12,000 children 2 to 5 years old demonstrated
be kept refrigerated (not frozen), and all four doses must be taken to 90% efficacy over a trial period of 4 years using recombinant exoprotein
achieve maximum efficacy. Each capsule should be taken with cool A of Pseudomonas aeruginosa (Vi-rEPA).434 Anti-Vi IgG titers were higher
liquid no warmer than 37°C (98.6°F) approximately 1 hour before a and longer lasting with conjugate Vi vaccines compared with Vi alone
meal. This regimen should be completed 1 week before potential exposure. in adults and children from endemic areas.631
The vaccine manufacturer recommends that Ty21a not be administered None of the typhoid vaccines approaches 100% efficacy, and a large
to infants or children younger than 6 years. inoculum of S. typhi can overcome vaccine-induced immunity. Vaccines
A parenteral subunit vaccine, ViCPS, was licensed in 1994 for use do not substitute for proper hygiene and appropriate food handling
in adults and children as young as 2 years of age. Primary vaccination practices.
with ViCPS consists of one 0.5-mL (25-µg) dose administered intra-
muscularly and should be given at least 2 weeks before potential exposure. Adverse Events
The manufacturer does not recommend the vaccine for children younger Reactions to the oral Ty21a vaccine usually are mild and consist of
than 2 years. transient gastrointestinal upset, fever, headache, and an occasionally
Various protein conjugated Vi polysaccharide vaccines have been rash. These reactions occur in less than 5% of recipients.366 Adverse
developed and are used in countries other than the United States, Two systemic effects are reported in less than 1% of vaccinees, and neither
were recently licensed in India for use in children 3 months or older.366,631 the development of bacteremia nor person-to-person transmission has
In circumstances of continued or repeated exposure to S. typhi, been reported.324
booster doses are recommended to maintain immunity after primary Reactions to the ViCPS vaccine also occur infrequently and are
immunization. The optimal booster schedule for either vaccine has not reported in approximately 7% of recipients366 These reactions include
been determined. Current recommendations for revaccination with tenderness at the injection site, erythema and induration, fever, and
either vaccine are provided in Table 245.20. No data have been reported rarely, rashes. Systemic complaints occur in less than 2%.324 In contrast,
comparing the use of one vaccine as a booster after primary immunization reactions to the inactivated, whole-cell vaccine occur more commonly
with the other. and are more severe; they include fever in as many as 24% of recipients,
headache, and severe local pain or swelling in as many as 35% of vac-
Efficacy cinees. Between 13% and 24% of vaccinees have missed school or work
Although field trials have demonstrated approximately 80% efficacy of because of adverse reactions.430
typhoid vaccines in US travelers,446 no comparative studies have been Reactions to the conjugate Vi vaccines were mild and limited to
performed.366 For the heat-phenol–inactivated, whole-cell vaccine, fever and mild erythema. There were no serious adverse events.434,631
efficacy ranged from 51% to 88% and lasted for as long as 7 years. The
oral, live attenuated Ty21a vaccine stimulates humoral (i.e., serum IgG Indications
and mucosal IgA antibodies) and cell-mediated immune responses. In Typhoid vaccination in the United States is recommended only for the
trials of the Ty21a vaccine, efficacy ranged from 42% to 96% after following groups366,503:
administration of the initial series of three doses, with the lower effica- • Travelers in areas where typhoid fever is endemic and for whom
cies seen in trials from areas with highly endemic disease.429,605 The the risk of exposure is recognized; risk of exposure is greatest with
optimal schedule for booster immunizations has not been determined travel to the Indian subcontinent, Latin America, Asia, the Middle
for this vaccine. Protective immunity persisted for at least 7 years in East, and Africa
field trials in endemic countries where herd immunity may have • People with intimate exposure to a documented S. typhi carrier, as
an effect.324 Revaccination is recommended with the same four-capsule occurs with continuing household contact
regimen every 5 years for as long as continued exposure is likely • Laboratory workers who have frequent contact with S. typhi and
to occur. people living in areas outside the United States with endemic typhoid
Immunization with ViCPS results in seroconversion, defined as a infection
fourfold rise in serum anti-Vi antibody, in 80% of vaccinees within 2 Vaccination is not recommended for people attending summer camp
weeks in endemic and nonendemic areas.324,342,397 Because the vaccine or for those in areas of natural disaster or for control of common-source
2594 PART V  Prevention of Infectious Diseases

outbreaks. Doses and schedules for the different typhoid vaccines are reported in vaccine recipients after the administration of more than
given in the recommendations of the CDC.366,503 540 million doses; 16 (89%) had received a vaccine dose within the
previous 10 years.617
Contraindications
Ty21a is a live attenuated vaccine and should not be given to immuno- Adverse Events
compromised patients, including those receiving high doses of The 17D-204 and 17DD yellow fever vaccines are among the safest and
corticosteroids and people with HIV infection.366,503 Ty21a vaccine also most effective viral vaccines.484 Since 1965, approximately 8 million
should not be administered during a gastrointestinal illness or concurrent doses of 17D-derived yellow fever vaccine have been administered to
with certain antibiotics. The 2015 Red Book recommends avoiding US travelers, and approximately 300 million doses have been administered
antimicrobial therapy for at least 24 hours before the first dose of oral to people in areas where yellow fever is endemic. Although 10% to 30%
typhoid vaccine through 7 days after the last dose.45 Reports are conflicting of people who receive vaccine report headaches, myalgia, and low-grade
regarding the use of the antimalarial agents atovaquone and proguanil; fever that begin within a few days after vaccination and last 5 to 10
however, chloroquine and mefloquine do not appear to interfere with days, less than 1% report temporarily curtailing their usual activities.
the immune response to oral Ty21a. Neither of the available typhoid Historically, yellow fever vaccine–associated adverse events were seen
vaccines should be given to anyone with a history of severe local or primarily among infants and manifested as encephalitis. However, with
systemic reactions after receiving a previous dose of vaccine. the current yellow fever vaccine, serious adverse events associated with
Ty21a is not approved for children younger than 6 years, and ViCPS vaccine rarely occur. A systematic review found that no serious adverse
is not approved for children younger than 2 years. The whole-cell, events were reported in four RCTs of infants and children (n = 1866).636
inactivated vaccine available outside of the United States is approved Three well-characterized serious adverse events may occur after
for use in children between 6 months and 2 years of age. yellow fever vaccine administration: immediate hypersensitivity or
Information is not available on the safety of these vaccines when anaphylactic reactions, yellow fever vaccine–associated neurologic disease
they are used during pregnancy. It is prudent on theoretical grounds (YEL-AND), and yellow fever vaccine–associated viscerotropic disease
to avoid vaccinating pregnant women. (YEL-AVD). Immediate hypersensitivity reactions, characterized by rash,
urticaria, or bronchospasm, or a combination of these features is
Yellow Fever Vaccine uncommon (1.8 cases/100,000 doses). Unrecognized allergy to eggs or
Yellow fever occurs only in sub-Saharan Africa and tropical South chicken or to the hydrolyzed gelatin used to stabilize the vaccine may
America, where it causes an estimated 200,000 cases and 30,000 deaths be responsible for hypersensitivity reactions.
annually.564 During the past 2 decades, 90% of all yellow fever cases YEL-AND is a serious but rarely fatal adverse event that occurs
were reported from countries in West Africa, where travelers are thought among people of all ages. YEL-AND manifests as several distinct clinical
to be at the highest risk of travel-associated yellow fever.617 Clinical syndromes, including meningoencephalitis (i.e., neurotropic disease),
disease ranges from a mild, nonspecific febrile illness to severe disease Guillain-Barré syndrome, acute disseminated encephalomyelitis (ADEM),
with jaundice and hemorrhage. and bulbar palsy, starting 3 to 28 days after vaccination, usually in
Yellow fever is an important vaccine-preventable disease among first-time recipients.140 Meningoencephalitis occurs as a result of direct
travelers to areas where yellow fever occurs. The yellow fever epidemic yellow fever vaccine viral invasion of the central nervous system (CNS)
zones include Bolivia, Brazil, Colombia, Ecuador, French Guiana, Guyana, with infection of the meninges or the brain, or both. The other neurologic
Paraguay, Peru, Suriname, Venezuela, and much of sub-Saharan Africa. syndromes (e.g., Guillain-Barré syndrome, ADEM) represent autoimmune
From 1970 to 2013, 10 cases of yellow fever were reported in unvaccinated manifestations in which antibodies or T cells produced in response to
travelers from the United States and Europe who traveled to West Africa the vaccine cross-react with neuronal epitopes and lead to central or
(five cases) or South America (five cases). The unvaccinated traveler’s peripheral nerve damage. The incidence of YEL-AND in the United
risk of acquiring yellow fever probably is increasing because potential States is 0.8 case per 100,000 doses.
zones of transmission of yellow fever are expanding to include urban In 2001, a previously unrecognized, potentially fatal adverse reaction
areas with large populations of susceptible humans and abundant among recipients of YF vaccine was first described. This syndrome
competent mosquito vectors. Vaccination is the most effective preventive previously was reported as febrile multiple organ dysfunction or failure
measure against yellow fever, a disease that has no specific treatment and now is called yellow fever vaccine–associated viscerotropic disease.
and may cause the death of 20% to 50% of patients.484 YEL-AVD mimics naturally acquired wild-type yellow fever disease,
with the vaccine virus proliferating and disseminating throughout the
Preparations host’s tissues. Since it was initially described, more than 65 confirmed
Yellow fever vaccines are derived from the original 17D yellow fever or suspected cases have been reported globally. On the basis of an
vaccine strain. The live attenuated 17D-204 and 17DD yellow fever analysis of cases for which information is available, YEL-AVD has
strains are the yellow fever vaccines most commonly used.484 The only occurred only after a recipient’s first yellow fever vaccination; no cases
yellow fever vaccine licensed in the United States is the 17D-204 vaccine, of YEL-AVD occurring in people receiving booster doses of the vaccine
which is prepared in chick embryos (YF-Vax, Sanofi Pasteur).618 have been reported. The median time from vaccination to disease onset
Primary immunization consists of a single, subcutaneous injection is approximately 4 days. The case-fatality ratio for reported cases is
of 0.5 mL of reconstituted, freeze-dried vaccine for adults and children. approximately 60%. The incidence of YEL-AVD in the United States is
The vaccine should be stored at 2°C to 8°C. Because the vaccine does 0.4 case per 100,000 doses.
not contain a preservative, reconstituted vaccine that remains unused Several risk estimates for YEL-AVD have been published. On the
after 1 hour must be properly disposed. basis of VAERS data, the reporting rate of YEL-AVD is 3 to 4 cases per
1 million doses distributed.435,462
Immunogenicity and Efficacy The recognition of these adverse events may be challenging because
Seroconversion rates of 93% have been documented for young children the neurologic syndromes (e.g., encephalitis, myelitis) can be difficult
receiving yellow fever vaccine.710 Thirteen observational studies showed to distinguish from bacterial meningitis, encephalitis, or malaria, and
a seropositivity rate of 92% at least 10 years after receiving the vaccine the viscerotropic syndrome can be difficult to distinguish from hemor-
and 80% at least 20 years after vaccination.617 Immunity develops by rhagic fevers, viral hepatitis, and many other causes of multisystem
the tenth day after primary vaccination. The titer of virus-neutralizing failure.
antibodies in the sera of vaccinees is used as a surrogate for vaccine
effectiveness. Indications
No human efficacy studies have been performed with yellow fever Yellow fever vaccine is recommended for people 9 months or older
vaccine. However, the number of yellow fever cases was substantially traveling to or residing in areas where yellow fever is endemic.618 Because
reduced after the introduction of the vaccine, supporting its protective of the increased risk of neurologic complications, infants 6 to 8 months
role in humans. Worldwide, only 18 cases of yellow fever have been of age should be considered for vaccination only when travel to high-risk
CHAPTER 245  Active Immunizing Agents 2595

areas is unavoidable and high-level protection against mosquito exposure immunosuppressive or immunomodulatory therapies (e.g., high-dose
is not feasible. Infants younger than 6 months have a substantially systemic corticosteroids, alkylating drugs, antimetabolites, tumor necrosis
increased risk of neurologic disease, and vaccination should not be factor-α inhibitors, interleukin-1 and interleukin-6 blocking agents,
given to this age group. Vaccination for international travel to certain other monoclonal antibodies such as rituximab and alemtuzumab) and
destinations is required. should be avoided in most cases according to the drug manufacturers.
Yellow fever vaccination requirements for specific countries are A history of thymectomy or indirect radiation therapy is not considered
available on the CDC Traveler’s Health website (http://wwwnc.cdc.gov/ a contraindication.
travel/yellowbook/2016/infectious-diseases-related-to-travel/yellow- Family members of immunosuppressed people, who themselves have
fever). To obtain an international certificate of vaccination, a yellow no contraindications, may receive yellow fever vaccine. If travel to an
fever vaccine approved by the WHO and administered at a designated epidemic or endemic area is necessary, the patient should be instructed
yellow fever vaccine center is required. Yellow fever vaccine centers in in ways to avoid mosquitoes and given a vaccine waiver letter.
the United States can be identified by contacting state or local health People with a history of systemic anaphylaxis to eggs or chicken
departments or visiting the CDC website (http://wwwnc.cdc.gov/travel/ should not be vaccinated because the vaccines contain egg proteins and
yellow-fever-vaccination-clinics/search). If the risks of potential serious on rare occasion may induce immediate allergic reactions. People may
adverse events caused by the yellow fever vaccine are considered greater also develop allergies to the vaccine because it contains gelatin and
than the potential benefits for exposed individuals, a medical waiver latex from the vial stopper. Less severe or local manifestations of allergy
letter can be provided, and guidance is provided on the CDC website to eggs or to feathers are not contraindications to yellow fever vaccine
(http://wwwnc.cdc.gov/travel/yellowbook/2016/infectious-diseases- administration and do not warrant performing vaccine skin testing.36
related-to-travel/yellow-fever#4731). If international quarantine regulations are the only reason to immunize
Historically, the WHO international health regulations required a patient known to be hypersensitive to eggs, a medical waiver letter
revaccination at 10-year intervals. However, evidence from several studies should be provided. If immunization of an individual with a questionable
indicated that yellow fever immunity persists for many decades. In history of egg hypersensitivity is considered essential because of the
2015, the CDC’s ACIP issued a recommendation that only a single dose high risk of exposure, an intradermal skin test can be given as directed
of yellow fever vaccine is required to provide long-lasting protection in the vaccine package insert.618
for most travelers except for pregnant women, hematopoietic stem cell
transplant recipients and HIV-infected people. The WHO has removed Vaccines Related to Bioterrorism
the booster requirement from the international health regulations as Anthrax and smallpox vaccines are potentially available to protect children
of 2016.617 against bioterrorist attacks with Bacillus anthracis and variola. Anthrax
Yellow fever vaccines can be administered at the same time as vaccine is not approved by the FDA for use in children. The AAP updated
inactivated vaccines and other live attenuated viral vaccines, except for its recommendations on exposure to anthrax through bioterrorism in
the MMR vaccine, because the immune responses against yellow fever, 2014.86 Until there are sufficient data to support FDA approval, anthrax
mumps, and rubella are inhibited.408 These vaccines should be given vaccine will be made available for children at the time of an event as
30 days apart. an investigational vaccine through an expedited process that requires
institutional review board approval, including the use of appropriate
Precautions and Contraindications consent documents. Information on the process required for use of
The risk of having adverse reactions appears to be age related.618 Infants anthrax vaccine in children will be available on the CDC website at the
younger than 9 months should not receive yellow fever vaccine because time of an event (https://www.cdc.gov/anthrax/).703
of the increased risk of vaccine-associated neurotropic disease developing The only smallpox vaccine licensed in the United States is ACAM2000,
in this age group.30 Immunization should be delayed until an infant is a live virus vaccine.153 In the absence of a smallpox outbreak, preexposure
at least 9 months of age. In unusual circumstances, physicians considering smallpox immunization is not recommended for children.46 Anthrax
vaccinating infants 6 to 8 months of age should contact the applicable and smallpox vaccines are reviewed in the relevant disease-specific
division of the CDC for advice (i.e., CDC Division of Vector-Borne chapters of this textbook.
Infectious Diseases or the CDC Division of Global Migration and
Quarantine: 800-232-4636). Investigational Vaccines
No major adverse effects of yellow fever vaccine on the developing Routine immunizations for children have virtually eliminated many
fetus have been demonstrated. A slight increased risk was observed for infectious diseases from the United States. These successes have encour-
minor, mostly skin, malformations in infants. A higher rate of spontane- aged research to develop vaccines to prevent other serious viral and
ous abortions in pregnant women receiving the vaccine was reported bacterial diseases affecting children. A 1985 report by the IOM of the
but not substantiated. Vaccine administration to pregnant women usually National Academy of Sciences reviewed the benefits that would be
is not indicated because the vaccine is a live virus. Pregnant women associated with the development and use of new and improved vaccines
should be considered for vaccination only when travel to high-risk in the United States.212 The report listed 14 diseases for which vaccines
areas is required and protection against mosquito exposure is not feasible. were desirable. A 1999 study by the IOM observed that considerable
Considering that immune responses to vaccination during pregnancy progress had been made since the 1985 study.213 Seven of 14 vaccines
may vary, serologic testing can be considered to document a protective listed in the 1985 study as domestic priorities for development are now
immune response to the vaccine. licensed. They include acellular pertussis vaccine, LAIV, and vaccines
Three YEL-AND cases have been reported in exclusively breastfed against hepatitis A and B, Hib, varicella, and rotavirus.
infants whose mothers were vaccinated with yellow fever vaccine. All The 1999 IOM report used a new quantitative model to compare
three infants were younger than 1 month and diagnosed with encephalitis. the cost and health benefits of developing candidate vaccines.213 This
Until more data are available, yellow fever vaccine should be avoided model can be used to evaluate the potential impact of a new vaccine
in breastfeeding women. However, when travel of breastfeeding mothers on public health. In the 1999 report, the model was used to evaluate
to a yellow fever–endemic area cannot be avoided, these women should diseases for which candidate vaccines were being developed. The report
be vaccinated. divided 26 candidate vaccines into four groups, arranged from most
Yellow fever vaccine, which is a live attenuated virus vaccine, poses to least favorable for development. The four vaccines in the top tier
a risk of developing encephalitis or other serious adverse events for include a cytomegalovirus vaccine given to adolescents, a universal
patients with conditions that commonly result in immunosuppression influenza vaccine, a group B Streptococcus vaccine for high-risk adults
(i.e. AIDS or symptomatic HIV infection, primary immunodeficiencies, and pregnant women, and an S. pneumoniae vaccine for infants and
thymic disease with abnormal immune function such as thymoma or seniors. Other diseases for which vaccines would be desirable included
myasthenia gravis, malignancy, transplantation, and radiation therapy). Chlamydia trachomatis, enterotoxigenic E. coli, Epstein-Barr virus,
The vaccine should be avoided by these people. Although not specifically Helicobacter pylori, hepatitis C virus, herpes simplex virus, human
tested, the vaccine is presumed to pose risks for patients receiving papilloma virus (HPV), M. tuberculosis, Neisseria gonorrhoeae, respiratory
2596 PART V  Prevention of Infectious Diseases

high public health importance, as identified by the WHO Product


BOX 245.2  Pathogens With Important Public Development for Vaccines Advisory Committee (PDVAC).687 PDVAC
Health Implications That Have No Licensed Vaccines is a body of independent experts that was established in 2014 to guide
Campylobacter jejuni the WHO and the vaccine development community along the pathway
Chikungunya virus toward the goal of licensure and deployment in countries with the
highest disease burden. These vaccines are listed in Box 245.2.
Dengue virus
Enterotoxigenic Escherichia coli
Enterovirus 71 (EV71) NEW REFERENCES SINCE THE SEVENTH EDITION
Group B Streptococcus (GBS) 1. Abu Raya B, Srugo I, Kessel A, et al. The decline of pertussis-specific antibodies
Herpes simplex virus after tetanus, diphtheria, and acellular pertussis immunization in late pregnancy.
J Infect Dis. 2015;212:1869-1873.
Human immunodeficiency virus type 1 (HIV-1) 4. Adams DA, Thomas KR, Jajosky RA, et al. Summary of notifiable infectious
Influenza virus (i.e., no universal vaccine) diseases and conditions—United States, 2014. MMWR Morb Mortal Wkly Rep.
Leishmania spp. (i.e., leishmaniasis) 2016;63:1-152.
Mycobacterium tuberculosis (i.e., tuberculosis) 5. Advisory Committee on Immunization Practices, Centers for Disease Control
and Prevention. Immunization of health-care personnel: recommendations of
Necator americanus and Ancylostoma duodenale (i.e., human hookworm
the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm
disease) Rep. 2011;60(RR-7):1-45.
Nipah virus 7. Albertson JP, Clegg WJ, Reid HD, et al. Mumps outbreak at a university and
Nontyphoidal Salmonella spp. (i.e., salmonellosis) recommendation for a third dose of measles-mumps-rubella vaccine—Illinois,
Norovirus 2015-2016. MMWR Morb Mortal Wkly Rep. 2016;65:731-734.
8. Ali M, Nelson AR, Lopez AL, et al. Updated global burden of cholera in endemic
Plasmodium spp. (i.e., malaria) countries. PLoS Negl Trop Dis. 2015;9:e0003832.
Respiratory syncytial virus (RSV) 9. Aliabadi N, Tate JE, Haynes AK, et al. Sustained decrease in laboratory detection
Salmonella enterica subspecies (i.e., paratyphoid fever) of rotavirus after implementation of routine vaccination—United States, 2000-
Schistosoma mansoni, S. haematobium, and S. japonicum (i.e., 2014. MMWR Morb Mortal Wkly Rep. 2015;64:337-342.
13. American Academy of Pediatrics Committee on Infectious Diseases. Updated rec-
schistosomiasis)
ommendations on the use of meningococcal vaccines. Pediatrics. 2014;134:400-403.
Shigella spp. (i.e., shigellosis) 16. American Academy of Pediatrics Committee on Infectious Diseases. Immunization
Staphylococcus aureus of adolescents: recommendations of the Advisory Committee on Immunization
Streptococcus pneumoniae Practices, the American Academy of Pediatrics, the American Academy of Family
Streptococcus pyogenes Physicians, and the American Medical Association. American Academy of Pediatrics
Committee on Infectious Diseases. Pediatrics. 1997;99:479-488.
Trypanosoma cruzi (i.e., Chagas disease) 28. American Academy of Pediatrics. Active immunization. In: Kimberlin DW,
Zika virus Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee
on Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of
Pediatrics; 2015:13-43.
29. American Academy of Pediatrics. Arboviruses. In: Kimberlin DW, Brady MT,
syncytial virus, parainfluenza virus, Shigella, and groups A and B Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infec-
tious Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics;
streptococci. Two vaccines listed in the 1999 study are now licensed:
2015:240-246.
pneumococcal conjugate vaccine for infants and seniors and HPV vaccine. 30. American Academy of Pediatrics. Committee on Native American Child Health
Progress in vaccine development is periodically reviewed and and Committee on Infectious Diseases. Immunizations for Native American
published by National Institute of Allergy and Infectious Diseases children. American Academy of Pediatrics. Pediatrics. 1999;104:564-567.
(NIAID) scientists in The Jordan Report.497 The 2012 report discusses 31. American Academy of Pediatrics. Diphtheria. In: Kimberlin DW, Brady MT, Jackson
progress in vaccine development and states that we now have licensed MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious Diseases.
vaccines against Hib and pneumococcal types that cause high childhood 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:325-329.
morbidity and mortality rates, against hepatitis A and B, against 32. American Academy of Pediatrics. infections. In: Kimberlin DW, Brady MT, Jackson
rotaviruses, and against varicella. We also have improved vaccines against MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious Diseases.
30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:368-376.
such diseases as influenza and pertussis.
33. American Academy of Pediatrics. Hepatitis A. In: Kimberlin DW, Brady MT, Jackson
Advances in biotechnology, increased understanding of the virulence MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious Diseases.
factors of infectious agents, and knowledge of the host immune response 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:391-399.
have led to new approaches for vaccine development.256 Technological 34. American Academy of Pediatrics. Hepatitis B. In: Kimberlin DW, Brady MT, Jackson
advances in making vaccines, including protein conjugation of bacterial MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious Diseases.
polysaccharides, DNA vaccines, viral chimeras, viral vectors, and other 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:400-423.
novel platforms have been developed. Vaccines are being developed by 35. American Academy of Pediatrics. Human papilloma viruses. In: Kimberlin DW,
the application of these newer technologies to numerous infectious Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee
agents for which effective vaccines are not currently available. The on Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of
Pediatrics; 2015:576-583.
development of vaccines for these pathogens requires solution of several
36. American Academy of Pediatrics. Immunization in special clinical circumstances.
immunologic problems, including pathogen variability, short effector In: Kimberlin DW, Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report
memory, evoking functional responses, and identification of antigens of the Committee on Infectious Diseases. 30th ed. Elk Grove Village, IL: American
that generate protective responses.539 Academy of Pediatrics; 2015:68-107.
The 2012 Jordan Report states that many challenges remain. Progress 37. American Academy of Pediatrics. Influenza. In: Kimberlin DW, Brady MT,
on vaccines to prevent some high-burden diseases has been frustratingly Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious
slow, although there is a growing pipeline of novel vaccines against Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:
dengue, malaria, tuberculosis, and others. The Report lists approximately 476-493.
600 vaccine candidates in development against an estimated 110 38. American Academy of Pediatrics. Measles. In: Kimberlin DW, Brady MT,
Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious
pathogens. Considering the resource constraints in vaccine development,
Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:
approaches that are most likely to produce high-priority vaccines need 535-547.
to be identified. 39. American Academy of Pediatrics. Meningococcal Infections. In: Kimberlin DW,
A special issue of Vaccine published in June 2016 focuses on the Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee
research and development pipeline of vaccines against 25 pathogens on Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of
for which no licensed vaccines currently exist but for which there is Pediatrics; 2015:547-558.
CHAPTER 245  Active Immunizing Agents 2597

40. American Academy of Pediatrics. Mumps. In: Kimberlin DW, Brady MT, Jackson 96. Bryant KA, Marshall GS, Marchant CD, et al. Immunogenicity and safety of H
MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious Diseases. influenzae type b–N meningitidis C/Y conjugate vaccine in infants. Pediatrics.
30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:564-568. 2011;127:e1375-e1385.
41. American Academy of Pediatrics. Pertussis (whooping cough). In: Kimberlin DW, 97. Buckle GC, Walker CL, Black RE. Typhoid fever and paratyphoid fever: systematic
Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee review to estimate global morbidity and mortality for 2010. J Glob Health.
on Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of 2012;2:010401.
Pediatrics; 2015:608-621. 98. Burns CC, Diop OM, Sutter RW, et al. Vaccine-derived polioviruses. J Infect Dis.
42. American Academy of Pediatrics. Pneumococcal infections. In: Kimberlin DW, 2014;210(suppl 1):S283-S293.
Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee 101. Byington CL, Maldonado YA, Barnett ED, et al. Recommendations for preven-
on Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of tion and control of influenza in children, 2016–2017. Pediatrics. 2016;138:pii:
Pediatrics; 2015:626-638. e20162527.
43. American Academy of Pediatrics. Poliovirus Infections. In: Kimberlin DW, 104. Castilla J, Garcia Cenoz M, Arriazu M, et al. Effectiveness of Jeryl Lynn-containing
Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee vaccine in Spanish children. Vaccine. 2009;27:2089-2093.
on Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of 105. Centers for Disease Control and Prevention. Addition of history of intussusception
Pediatrics; 2015:644-650. as a contraindication for rotavirus vaccination. MMWR Morb Mortal Wkly Rep.
44. American Academy of Pediatrics. Rubella. In: Kimberlin DW, Brady MT, Jackson 2011;60:1427.
MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious Diseases. 111. Centers for Disease Control and Prevention. Estimates of deaths associated with
30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:688-695. seasonal influenza—United States, 1976-2007. MMWR Morb Mortal Wkly Rep.
45. American Academy of Pediatrics. Salmonella Infections. In: Kimberlin DW, 2010;59:1057-1062.
Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee 121. Centers for Disease Control and Prevention. Licensure of a 13-valent pneu-
on Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of mococcal conjugate vaccine (PCV13) and recommendations for use among
Pediatrics; 2015:695-702. children—Advisory Committee on Immunization Practices (ACIP), 2010. MMWR
46. American Academy of Pediatrics. Smallpox (variola). In: Kimberlin DW, Brady Morb Mortal Wkly Rep. 2010;59:258-261.
MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on 125. Centers for Disease Control and Prevention. Mumps outbreak on a univer-
Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; sity campus—California, 2011. MMWR Morb Mortal Wkly Rep. 2012;61:
2015:709-712. 986-989.
47. American Academy of Pediatrics. Tetanus (Lockjaw). In: Kimberlin DW, Brady 129. Centers for Disease Control and Prevention. Notice to readers: newly licensed
MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on smallpox vaccine to replace old smallpox vaccine. MMWR Morb Mortal Wkly
Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; Rep. 2008;57:207-208.
2015:773-778. 132. Centers for Disease Control and Prevention. Pertussis—United States, 1997-2000.
48. American Academy of Pediatrics. Tuberculosis. In: Kimberlin DW, Brady MT, MMWR. 2002;51:73-76.
Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infec- 135. Centers for Disease Control and Prevention. Poliomyelitis prevention in the
tious Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; United States: introduction of a sequential vaccination schedule of inactivated
2015:805-831. poliovirus vaccine followed by oral poliovirus vaccine. Recommendations of
49. American Academy of Pediatrics. Varicella-zoster. In: Kimberlin DW, Brady MT, the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm
Jackson MA, Long SS, eds. Red Book: 2012 Report of the Committee on Infec- Rep. 1997;46(RR-3):1-25.
tious Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 138. Centers for Disease Control and Prevention. Progress in introduction of pneu-
2015:846-860. mococcal conjugate vaccine—worldwide, 2000-2012. MMWR Morb Mortal Wkly
50. Amirthalingam G, Andrews N, Campbell H, et al. Effectiveness of maternal Rep. 2013;62:308-311.
pertussis vaccination in England: an observational study. Lancet. 2014;384(9953): 145. Centers for Disease Control and Prevention. Rubella and congenital rubella
1521-1528. syndrome—United States, January 1, 1991-May 7, 1994. MMWR Morb Mortal
60. Atwell JE, Van Otterloo J, Zipprich J, et al. Nonmedical vaccine exemptions and Wkly Rep. 1994;43:391, 397-401.
pertussis in California, 2010. Pediatrics. 2013;132:624-630. 146. Centers for Disease Control and Prevention. Seasonal influenza vaccine safety: a
67. Barskey AE, Glasser JW, LeBaron CW. Mumps resurgences in the United summary for clinicians, 2016. https://www.cdc.gov/flu/professionals/vaccination/
States: a historical perspective on unexpected elements. Vaccine. 2009;27: vaccine_safety.htm.
6186-6195. 147. Centers for Disease Control and Prevention. Syncope after vaccination—United
68. Baylis SA, Finsterbusch T, Bannert N, et al. Analysis of porcine circovirus type States, January 2005-July 2007. MMWR Morb Mortal Wkly Rep. 2008;57:
1 detected in Rotarix vaccine. Vaccine. 2011;29:690-697. 457-460.
69. Beasley RP, Detels R, Kim KS, et al. Prevention of rubella during an epidemic 148. Centers for Disease Control and Prevention. Tetanus surveillance—United States,
on Taiwan. HPV-77 and RA 27-3 rubella vaccines administered subcutaneously 2001-2008. MMWR Morb Mortal Wkly Rep. 2011;60:365-369.
and intranasally HPV-77 vaccine mixed with mumps and/or measles vaccines. 161. Centers for Disease Control and Prevention. Updated recommendations for
Am J Dis Child. 1969;118:301-306. use of tetanus toxoid, reduced diphtheria toxoid, and acellular pertussis vaccine
73. Belongia EA, Irving SA, Shui IM, et al. Real-time surveillance to assess risk (Tdap) in pregnant women—Advisory Committee on Immunization Practices
of intussusception and other adverse events after pentavalent, bovine-derived (ACIP), 2012. MMWR Morb Mortal Wkly Rep. 2013;62:131-135.
rotavirus vaccine. Pediatr Infect Dis J. 2010;29:1-5. 163. Centers for Disease Control and Prevention. Use of 13-valent pneumococcal
79. Black CL, Yue X, Ball SW, et al. Influenza vaccination coverage among health conjugate vaccine and 23-valent pneumococcal polysaccharide vaccine among
care personnel—United States, 2014-15 influenza season. MMWR Morb Mortal children aged 6-18 years with immunocompromising conditions: recommenda-
Wkly Rep. 2015;64:993-999. tions of the Advisory Committee on Immunization Practices (ACIP). MMWR
80. Black S, Greenberg DP. A combined diphtheria, tetanus, five-component acellular Morb Mortal Wkly Rep. 2013;62:521-524.
pertussis, poliovirus and Haemophilus influenzae type b vaccine. Expert Rev 166. Centers for Disease Control and Prevention. Use of Japanese encephalitis vaccine
Vaccines. 2005;4:793-805. in children: recommendations of the advisory committee on immunization
85. Borgono JM, Greiber R, Solari G, et al. A field trial of combined measles-mumps- practices, 2013. MMWR Morb Mortal Wkly Rep. 2013;62:898-900.
rubella vaccine. Satisfactory immunization with 188 children in Chile. Clin Pediatr 170. Centers for Disease Control. Mumps prevention. MMWR Morb Mortal Wkly
(Phila). 1973;12:170-172. Rep. 1989;38:388-392, 397-400.
86. Bradley JS, Peacock G, Krug SE, et al. Pediatric anthrax clinical management. 171. Centers for Disease Control. Mumps—United States, 1985-1988. MMWR Morb
Pediatrics. 2014;133:e1411-e1436. Mortal Wkly Rep. 1989;38:101-105.
87. Brady MT, Byington CL, Davies HD, et al. Immunization for Streptococcus 172. Centers for Disease Control. Pneumococcal polysaccharide vaccine: Recom-
pneumoniae infections in high-risk children. Pediatrics. 2014;134:1230-1233. mendations of the Advisory Committee on Immunization Practices (ACIP).
89. Briere EC, Rubin L, Moro PL, et al. Prevention and control of Haemophilus MMWR. 1989;38:64-68, 73-76.
influenzae type b disease: recommendations of the advisory committee on 175. Centers for Disease Control. Rubella and congenital rubella—United States,
immunization practices (ACIP). MMWR Recomm Rep. 2014;63(RR-01):1-14. 1983. MMWR Morb Mortal Wkly Rep. 1984;33:237-242, 247.
92. Broliden K, Abreu ER, Arneborn M, et al. Immunity to mumps before and after 178. Centers for Disease Control. Surveillance of tetanus, 1987 and 1988. Wkly Epidemiol
MMR vaccination at 12 years of age in the first generation offered the two-dose Rec. 1990;65:261-263.
immunization programme. Vaccine. 1998;16:323-327. 180. Chaiken BP, Williams NM, Preblud SR, et al. The effect of a school entry law
93. Brooks R, Woods CW, Benjamin DK Jr, et al. Increased case-fatality rate on mumps activity in a school district. JAMA. 1987;257:2455-2458.
associated with outbreaks of Neisseria meningitidis infection, compared with 181. Chamot E, Toscani L, Egger P, et al. Estimation of the efficacy of three strains
sporadic meningococcal disease, in the United States, 1994-2002. Clin Infect of mumps vaccines during an epidemic of mumps in the Geneva canton
Dis. 2006;43:49-54. (Switzerland). Rev Epidemiol Sante Publique. 1998;46:100-107.
2598 PART V  Prevention of Infectious Diseases

183. Cheek JE, Baron R, Atlas H, et al. Mumps outbreak in a highly vaccinated school 256. Ellis RW, Rappuoli R, Ahmed S. New technologies for making vaccines. In: Plotkin
population. Evidence for large-scale vaccination failure. Arch Pediatr Adolesc S, Orenstein W, Offit P, eds. Vaccines. 6th ed. Philadelphia, PA: Saunders-Elsevier;
Med. 1995;149:774-778. 2013:1182-1199.
187. Chen WH, Cohen MB, Kirkpatrick BD, et al. Single-dose live oral cholera vaccine 263. Feikin DR, Kagucia EW, Loo JD, et al. Serotype-specific changes in invasive
CVD 103-HgR protects against human experimental infection with Vibrio cholerae pneumococcal disease after pneumococcal conjugate vaccine introduction: a
O1 El Tor. Clin Infect Dis. 2016;62:1329-1335. pooled analysis of multiple surveillance sites. PLoS Med. 2013;10:e1001517.
188. Cherry JD, Brunell PA, Golden GS, et al. Report of the task force on pertussis 264. Feiterna-Sperling C, Bronnimann R, Tischer A, et al. Open randomized trial
and pertussis immunization—1988. Pediatrics. 1988;81:933-984. comparing the immunogenicity and safety of a new measles-mumps-rubella
189. Chesson HW, Dunne EF, Hariri S, et al. The estimated lifetime probability vaccine and a licensed vaccine in 12- to 24-month-old children. Pediatr Infect
of acquiring human papillomavirus in the United States. Sex Transm Dis. Dis J. 2005;24:1083-1088.
2014;41:660-664. 265. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality
190. Christenson B, Bottiger M. Long-term follow-up study of rubella antibodies in worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J
naturally immune and vaccinated young adults. Vaccine. 1994;12:41-45. Cancer. 2015;136:E359-E386.
192. Chung JR, Flannery B, Thompson MG, et al. Seasonal effectiveness of live 266. Fiebelkorn A, Barskey A, Hickman C, et al. Mumps. In: Prevention CfDCa, ed.
attenuated and inactivated influenza vaccine. Pediatrics. 2016;137:e20153279. Manual for the Surveillance of Vaccine-Preventable Diseases. 5th ed. Atlanta, GA:
198. Cochi SL, Broome CV, Hightower AW. Immunization of US children with US Department of Health and Human Services, Centers for Disease Control
Hemophilus influenzae type b polysaccharide vaccine. A cost-effectiveness model and Prevention; 2012.
of strategy assessment. JAMA. 1985;253:521-529. 267. Fiebelkorn AP, Redd SB, Gastanaduy PA, et al. A comparison of postelimination
199. Cochi SL, Preblud SR, Orenstein WA. Prespectives on the relative resurgence of measles epidemiology in the United States, 2009-2014 versus 2001-2008. J Pediatric
mumps in the United States. Am J Dis Child. 1988;142:499-507. Infect Dis Soc. 2017;6:40-48.
203. Cohn A Proceedings of the Advisory Committee on Immunization Practices 274. Fletcher HA, Schrager L. TB vaccine development and the End TB Strategy:
(ACIP) meeting. Paper presented at the Advisory Committee on Immunization importance and current status. Trans R Soc Trop Med Hyg. 2016;110:212-218.
Practices (ACIP) meeting; 2010; Atlanta, GA. 275. Fogel A, Moshkowitz A, Rannon L, et al. Comparative trials of RA 27-3 and
204. Cohn AC, MacNeil JR, Clark TA, et al. Prevention and control of meningococcal Cendehill rubella vaccines in adult and adolescent females. Am J Epidemiol.
disease: recommendations of the Advisory Committee on Immunization Practices 1971;93:392-398.
(ACIP). MMWR Recomm Rep. 2013;62(RR-2):1-28. 276. Folaranmi T, Rubin L, Martin SW, et al. Use of serogroup B meningococcal vaccines
205. Cohn AC, MacNeil JR, Harrison LH, et al. Changes in Neisseria meningitidis in persons aged ≥10 years at increased risk for serogroup B meningococcal disease:
disease epidemiology in the United States, 1998-2007: implications for prevention recommendations of the Advisory Committee on Immunization Practices, 2015.
of meningococcal disease. Clin Infect Dis. 2010;50:184-191. MMWR Morb Mortal Wkly Rep. 2015;64:608-612.
209. Committee on Infectious Diseases, American Academy of Pediatrics. Recommended 277. Food and Drug Administration. December 10, 2014 approval letter—Gardasil 9.
childhood and adolescent immunization schedule—United States, 2016. Pediatrics. Silver Spring, MD: US Department of Health and Human Services; 2014.
2016;137:e20154531. 278. Food and Drug Administration. October 7, 2016 approval letter for human papil-
210. Committee on Infectious Diseases, American Academy of Pediatrics. HPV vaccine lomavirus 9-valent vaccine, recombinant to include a 2-dose regimen for individuals
recommendations. Pediatrics. 2012;129:602-605. 9 through 14 years of age. Silver Spring, MD: US Department of Health and
211. Committee on Infectious Diseases, American Academy of Pediatrics. Recom- Human Services; 2016.
mendations for serogroup B meningococcal vaccine for persons 10 years and 279. Fowlkes A, Steffens A, Temte J, et al. Incidence of medically attended influenza
older. Pediatrics. 2016;138:pii: e20161890. during pandemic and post-pandemic seasons through the Influenza Incidence
214. Committee to Study Priorities for Vaccine Development, Institute of Medicine. Surveillance Project, 2009-13. Lancet Respir Med. 2015;3:709-718.
Vaccines for the 21st Century: A Tool for Decision Making. Washington. DC: 281. Freestone DS, Reynolds GM, McKinnon JA, et al. Vaccination of schoolgirls
National Academies Press; 2000. against rubella. Assessment of serological status and a comparative trial of Wistar
215. Cortes JE, Curns AT, Tate JE, et al. Rotavirus vaccine and health care utilization RA 27/3 and Cendehill strain live attenuated rubella vaccines in 13-year-old
for diarrhea in U.S. Children. N Engl J Med. 2011;365:1108-1117. schoolgirls in Dudley. Br J Prev Soc Med. 1975;29:258-261.
216. Cortese MM, Jordan HT, Curns AT, et al. Mumps vaccine performance among 282. Fritzell B, Plotkin S. Efficacy and safety of a Haemophilus influenzae type b
university students during a mumps outbreak. Clin Infect Dis. 2008;46:1172-1180. capsular polysaccharide-tetanus protein conjugate vaccine. J Pediatr. 1992;121:
218. Coulehan JL, Michaels RH, Hallowell C, et al. Epidemiology of Haemophilus 355-362.
influenzae type B disease among Navajo Indians. Public Health Rep. 1984;99: 283. Furukawa T, Miyata T, Kondo K, et al. Rubella vaccination during an epidemic.
404-409. JAMA. 1970;213:987-990.
225. D’Amelio R, Biselli R, Fascia G, et al. Measles-mumps-rubella vaccine in the 284. Gaglani M, Pruszynski J, Murthy K, et al. Influenza vaccine effectiveness against
Italian armed forces. JAMA. 2000;284:2059. 2009 pandemic influenza A(H1N1) virus differed by vaccine type during 2013-2014
229. Dayan GH, Quinlisk MP, Parker AA, et al. Recent resurgence of mumps in the in the United States. J Infect Dis. 2016;213:1546-1556.
United States. N Engl J Med. 2008;358:1580-1589. 287. Gastanaduy PA, Redd SB, Fiebelkorn AP, et al. Measles—United States, January
231. Decker MD, Edwards KM. Haemophilus influenzae type b vaccines: history, choice 1-May 23, 2014. MMWR Morb Mortal Wkly Rep. 2014;63:496-499.
and comparisons. Pediatr Infect Dis J. 1998;17(suppl):S113-S116. 288. Gatchalian S, Cordero-Yap L, Lu-Fong M, et al. A randomized comparative trial
233. Deeks SL, Lim GH, Simpson MA, et al. An assessment of mumps vaccine in order to assess the reactogenicity and immunogenicity of a new measles
effectiveness by dose during an outbreak in Canada. CMAJ. 2011;183:1014-1020. mumps rubella (MMR) vaccine when given as a first dose at 12-24 months of
237. Dennehy PH, Vesikari T, Matson DO, et al. Efficacy of the pentavalent rotavirus age. Southeast Asian J Trop Med Public Health. 1999;30:511-517.
vaccine, RotaTeq(R) (RV5), between doses of a 3-dose series and with less than 295. Giebink GS. Preventing pneumococcal disease in children: recommendations
3 doses (incomplete regimen). Hum Vaccin. 2011;7:563-568. for using pneumococcal vaccine. Pediatr Infect Dis. 1985;4:343-348.
239. D’Hondt E. Possible approaches to develop vaccines against hepatitis A. Vaccine. 296. Gill CJ, Baxter R, Anemona A, et al. Persistence of immune responses after a
1992;10:S48-S52. single dose of Novartis meningococcal serogroup A, C, W-135 and Y CRM-197
241. Dominguez A, Torner N, Castilla J, et al. Mumps vaccine effectiveness in highly conjugate vaccine (Menveo® or Menactra®) among healthy adolescents. Hum
immunized populations. Vaccine. 2010;28:3567-3570. Vaccin. 2010;6:881-887.
243. Donegan K, King B, Bryan P. Safety of pertussis vaccination in pregnant women 304. Goldschneider I, Gotschlich EC, Artenstein MS. Human immunity to the
in UK: observational study. BMJ. 2014;349:g4219. meningococcus. I. The role of humoral antibodies. J Exp Med. 1969;129:
244. Donovan B, Franklin N, Guy R, et al. Quadrivalent human papillomavirus vac- 1307-1326.
cination and trends in genital warts in Australia: analysis of national sentinel 306. Gothefors L, Bergstrom E, Backman M. Immunogenicity and reactogenicity of
surveillance data. Lancet Infect Dis. 2011;11:39-44. a new measles, mumps and rubella vaccine when administered as a second dose
245. Donovan B, Grulich AE. The quadrivalent HPV vaccine is effective prophylaxis at 12 y of age. Scand J Infect Dis. 2001;33:545-549.
against HPV-related external genital lesions in young men. Evid Based Med. 307. Gotschlich EC, Goldschneider I, Artenstein MS. Human immunity to the
2011;16:157-158. meningococcus. IV. Immunogenicity of group A and group C meningococcal
246. Douvoyiannis M, Belamarich PF, Goldman DL. Tetanus after vaccine refusal polysaccharides in human volunteers. J Exp Med. 1969;129:1367-1384.
and an opportunity for the pediatric infectious diseases specialist. Clin Pediatr 308. Grassly NC. Immunogenicity and effectiveness of routine immunization with 1
(Phila). 2015;54:513-516. or 2 doses of inactivated poliovirus vaccine: systematic review and meta-analysis.
249. Dunne EF, Nielson CM, Stone KM, et al. Prevalence of HPV infection among J Infect Dis. 2014;210(suppl 1):S439-S446.
men: a systematic review of the literature. J Infect Dis. 2006;194:1044-1057. 314. Grillner L. Neutralizing antibodies after rubella vaccination of newly delivered
254. Ehrenkranz NJ, Ventura AK, Medler EM, et al. Clinical evaluation of a new women: a comparison between three vaccines. Scand J Infect Dis. 1975;7:169-172.
measles-mumps-rubella combined live virus vaccine in the Dominican Republic. 315. Grohskopf LA, Sokolow LZ, Broder KR, et al. Prevention and control of seasonal
Bull World Health Organ. 1975;52:81-85. influenza with vaccines. MMWR Recomm Rep. 2016;65:1-54.
CHAPTER 245  Active Immunizing Agents 2599

318. Group GA. Expanded programme on immunization. Global Advisory Group—Part 399. Kobayashi M, Bennett NM, Gierke R, et al. Intervals Between PCV13 and PPSV23
II. Achieving the major disease control goals. Wkly Epidemiol Rec. 1994;69:29-31, Vaccines: recommendations of the Advisory Committee on Immunization Practices
34-35. (ACIP). MMWR Morb Mortal Wkly Rep. 2015;64:944-947.
319. Guerra FA, Blatter MM, Greenberg DP, et al. Safety and immunogenicity of a 404. Kremer JR, Schneider F, Muller CP. Waning antibodies in measles and rubella
pentavalent vaccine compared with separate administration of licensed equivalent vaccinees—a longitudinal study. Vaccine. 2006;24:2594-2601.
vaccines in US infants and toddlers and persistence of antibodies before a preschool 406. Kristiansen PA, Jorgensen HJ, Caugant DA. Serogroup A meningococcal conjugate
booster dose: a randomized, clinical trial. Pediatrics. 2009;123:301-312. vaccines in Africa. Expert Rev Vaccines. 2015;14:1441-1458.
325. Haber P, Patel M, Izurieta HS, et al. Postlicensure monitoring of intussusception 412. Kuehn BM. FDA: Benefits of rotavirus vaccination outweigh potential contamina-
after RotaTeq vaccination in the United States, February 1, 2006, to September tion risk. JAMA. 2010;304:30-31.
25, 2007. Pediatrics. 2008;121:1206-1212. 420. Le Bouvier GL, Plotkin SA. Precipitin responses to rubella vaccine RA 27-3. J
331. Hampton LM, Farley MM, Schaffner W, et al. Prevention of antibiotic- Infect Dis. 1971;123:220-223.
nonsusceptible Streptococcus pneumoniae with conjugate vaccines. J Infect Dis. 422. LeBaron CW, Forghani B, Beck C, et al. Persistence of mumps antibodies after
2012;205:401-411. 2 doses of measles-mumps-rubella vaccine. J Infect Dis. 2009;199:552-560.
332. Hanna-Wakim R, Yasukawa LL, Sung P, et al. Immune responses to mumps 423. LeBaron CW, Forghani B, Matter L, et al. Persistence of rubella antibodies after
vaccine in adults who were vaccinated in childhood. J Infect Dis. 2008;197: 2 doses of measles-mumps-rubella vaccine. J Infect Dis. 2009;200:888-899.
1669-1675. 424. Lee SH, Ewert DP, Frederick PD, et al. Resurgence of congenital rubella syndrome
341. Hersh BS, Fine PE, Kent WK, et al. Mumps outbreak in a highly vaccinated in the 1990s. Report on missed opportunities and failed prevention policies
population. J Pediatr. 1991;119:187-193. among women of childbearing age. JAMA. 1992;267:2616-2620.
345. Hill HA, Elam-Evans LD, Yankey D, et al. Vaccination coverage among children 427. Leshem E, Moritz RE, Curns AT, et al. Rotavirus vaccines and health care utilization
aged 19-35 months—United States, 2015. MMWR Morb Mortal Wkly Rep. for diarrhea in the United States (2007-2011. Pediatrics. 2014;134:15-23.
2016;65:1065-1071. 431. Lewis JE, Chernesky MA, Rawls ML, et al. Epidemic of mumps in a partially
346. Hillary IB, Griffith AH. Persistence of rubella antibodies 15 years after subcutaneous immune population. Can Med Assoc J. 1979;121:751-754.
administration of Wistar 27/3 strain live attenuated rubella virus vaccine. Vaccine. 434. Lin FY, Ho VA, Khiem HB, et al. The efficacy of a Salmonella typhi Vi conjugate
1984;2:274-276. vaccine in two-to-five-year-old children. N Engl J Med. 2001;344:1263-1269.
347. Hilleman MR, Buynak EB, Weibel RE, et al. Live, attenuated mumps-virus vaccine. 436. Loharikar A, Newton AE, Stroika S, et al. Cholera in the United States, 2001-
N Engl J Med. 1968;278:227-232. 2011: a reflection of patterns of global epidemiology and travel. Epidemiol Infect.
348. Hilleman MR, Weibel RE, Buynak EB, et al. Live attenuated mumps-virus 2015;143:695-703.
vaccine. IV. Protective efficacy as measured in a field evaluation. N Engl J Med. 442. MacNeil JR, Bennett N, Farley MM, et al. Epidemiology of infant meningococcal
1967;276:252-258. disease in the United States, 2006-2012. Pediatrics. 2015;135:e305-e311.
352. Holmes SJ, Fritzell B, Guito KP, et al. Immunogenicity of Haemophilus influenzae 443. Macneil JR, Cohn AC, Zell ER, et al. Early estimate of the effectiveness of quad-
type b polysaccharide-tetanus toxoid conjugate vaccine in infants. Am J Dis rivalent meningococcal conjugate vaccine. Pediatr Infect Dis J. 2011;30:451-455.
Child. 1993;147:832-836. 444. MacNeil JR, Rubin L, Folaranmi T, et al. Use of serogroup B meningococcal
353. Horstmann DM, Schluederberg A, Emmons JE, et al. Persistence of vaccine-induced vaccines in adolescents and young adults: recommendations of the Advisory
immune responses to rubella: comparison with natural infection. Rev Infect Dis. Committee on Immunization Practices, 2015. MMWR Morb Mortal Wkly Rep.
1985;7(suppl 1):S80-S85. 2015;64:1171-1176.
354. Howson CP, Katz M, Johnston RB Jr, et al. Chronic arthritis after rubella vac- 445. MacNeil JR, Rubin LG, Patton M, et al. Recommendations for use of meningococcal
cination. Clin Infect Dis. 1992;15:307-312. conjugate vaccines in HIV-infected persons—Advisory Committee on Immuniza-
355. Hsieh YC, Wu FT, Hsiung CA, et al. Comparison of virus shedding after tion Practices, 2016. MMWR Morb Mortal Wkly Rep. 2016;65:1189-1194.
lived attenuated and pentavalent reassortant rotavirus vaccine. Vaccine. 446. Mahon BE, Newton AE, Mintz ED. Effectiveness of typhoid vaccination in US
2014;32:1199-1204. travelers. Vaccine. 2014;32:3577-3579.
366. Jackson BR, Iqbal S, Mahon B, et al. Updated recommendations for the use 452. Marin M, Quinlisk P, Shimabukuro T, et al. Mumps vaccination coverage and
of typhoid vaccine—Advisory Committee on Immunization Practices, United vaccine effectiveness in a large outbreak among college students—Iowa, 2006.
States, 2015. MMWR Morb Mortal Wkly Rep. 2015;64:305-308. Vaccine. 2008;26:3601-3607.
368. Jafri RZ, Ali A, Messonnier NE, et al. Global epidemiology of invasive meningococ- 453. Marin M, Willis ED, Marko A, et al. Closure of varicella-zoster virus-containing
cal disease. Popul Health Metr. 2013;11:17. vaccines pregnancy registry—United States, 2013. MMWR Morb Mortal Wkly
372. Johnson CE, Kumar ML, Whitwell JK, et al. Antibody persistence after primary Rep. 2014;63:732-733.
measles-mumps-rubella vaccine and response to a second dose given at four to 456. Markowitz LE, Dunne EF, Saraiya M, et al. Human papillomavirus vaccination:
six vs. eleven to thirteen years. Pediatr Infect Dis J. 1996;15:687-692. recommendations of the Advisory Committee on Immunization Practices (ACIP).
376. Johnson MG, Bradley KK, Mendus S, et al. Vaccine-preventable disease MMWR Recomm Rep. 2014;63(RR-05):1-30.
among homeschooled children: two cases of tetanus in Oklahoma. Pediatrics. 458. Markowitz LE, Preblud SR, Fine PE, et al. Duration of live measles vaccine-induced
2013;132:e1686-e1689. immunity. Pediatr Infect Dis J. 1990;9:101-110.
377. Jokinen S, Osterlund P, Julkunen I, et al. Cellular immunity to mumps virus 460. Marshall GS, Marchant CD, Blatter M, et al. Co-administration of a novel
in young adults 21 years after measles-mumps-rubella vaccination. J Infect Dis. Haemophilus influenzae type b and Neisseria meningitidis serogroups C and
2007;196:861-867. Y-tetanus toxoid conjugate vaccine does not interfere with the immune response
378. Joura EA, Giuliano AR, Iversen OE, et al. A 9-valent HPV vaccine against to antigens contained in infant vaccines routinely used in the United States.
infection and intraepithelial neoplasia in women. N Engl J Med. 2015;372: Hum Vaccin. 2011;7:258-264.
711-723. 461. Marshall HS, Richmond PC, Nissen MD, et al. A phase 2 open-label safety and
379. Kahn JA, Widdice LE, Ding L, et al. Substantial decline in vaccine-type human immunogenicity study of a meningococcal B bivalent rLP2086 vaccine in healthy
papillomavirus (HPV) among vaccinated young women during the first 8 adults. Vaccine. 2013;31:1569-1575.
years after HPV vaccine introduction in a community. Clin Infect Dis. 2016;63: 463. Martinon-Torres F, Czajka H, Center KJ, et al. 13-valent pneumococcal conjugate
1281-1287. vaccine (PCV13) in preterm versus term infants. Pediatrics. 2015;135:e876-e886.
388. Keyserling H, Papa T, Koranyi K, et al. Safety, immunogenicity, and immune 467. Mazurek GH, Jereb J, Vernon A, et al. Updated guidelines for using interferon
memory of a novel meningococcal (groups A, C, Y, and W-135) polysaccharide gamma release assays to detect Mycobacterium tuberculosis infection—United
diphtheria toxoid conjugate vaccine (MCV-4) in healthy adolescents. Arch Pediatr States, 2010. MMWR Recomm Rep. 2010;59(RR-5):1-25.
Adolesc Med. 2005;159:907-913. 469. McClenahan SD, Krause PR, Uhlenhaut C. Molecular and infectivity studies of
389. Kim-Farley R, Bart S, Stetler H, et al. Clinical mumps vaccine efficacy. Am J porcine circovirus in vaccines. Vaccine. 2011;29:4745-4753.
Epidemiol. 1985;121:593-597. 471. McLean HQ, Fiebelkorn AP, Temte JL, et al. Prevention of measles, rubella,
391. Kimura A, Toneatto D, Kleinschmidt A, et al. Immunogenicity and safety of congenital rubella syndrome, and mumps, 2013: summary recommendations of
a multicomponent meningococcal serogroup B vaccine and a quadrivalent the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm
meningococcal CRM197 conjugate vaccine against serogroups A, C, W-135, and Rep. 2013;62(RR-04):1-34.
Y in adults who are at increased risk for occupational exposure to meningococcal 474. Meites E, Kempe A, Markowitz LE. Use of a 2-dose schedule for human
isolates. Clin Vaccine Immunol. 2011;18:483-486. papillomavirus vaccination—updated recommendations of the Advisory Com-
393. Klein NP, Bartlett J, Fireman B, et al. Waning Tdap effectiveness in adolescents. mittee on Immunization Practices. MMWR Morb Mortal Wkly Rep. 2016;65:
Pediatrics. 2016;137:e20153326. 1405-1408.
396. Klinge J, Lugauer S, Korn K, et al. Comparison of immunogenicity and 475. Mellinger AK, Cragan JD, Atkinson WL, et al. High incidence of congenital
reactogenicity of a measles, mumps and rubella (MMR) vaccine in German rubella syndrome after a rubella outbreak. Pediatr Infect Dis J. 1995;14:573-578.
children vaccinated at 9-11, 12-14 or 15-17 months of age. Vaccine. 2000;18: 485. Monroe BP, Yager P, Blanton J, et al. Rabies surveillance in the United States
3134-3140. during 2014. J Am Vet Med Assoc. 2016;248:777-788.
2600 PART V  Prevention of Infectious Diseases

487. Moore MR, Link-Gelles R, Schaffner W, et al. Effect of use of 13-valent pneumococ- 550. Reef SE, Redd SB, Abernathy E, et al. Evidence used to support the achievement
cal conjugate vaccine in children on invasive pneumococcal disease in children and maintenance of elimination of rubella and congenital rubella syndrome in
and adults in the USA: analysis of multisite, population-based surveillance. Lancet the United States. J Infect Dis. 2011;204(suppl 2):S593-S597.
Infect Dis. 2015;15:301-309. 556. Richmond PC, Marshall HS, Nissen MD, et al. Safety, immunogenicity, and
488. Morgan JL, Baggari SR, McIntire DD, et al. Pregnancy outcomes after antepar- tolerability of meningococcal serogroup B bivalent recombinant lipoprotein 2086
tum tetanus, diphtheria, and acellular pertussis vaccination. Obstet Gynecol. vaccine in healthy adolescents: a randomised, single-blind, placebo-controlled,
2015;125:1433-1438. phase 2 trial. Lancet Infect Dis. 2012;12:597-607.
489. Moro PL, Cragan J, Tepper N, et al. Enhanced surveillance of tetanus toxoid, 559. Rivera L, Pena LM, Stainier I, et al. Horizontal transmission of a human rotavirus
reduced diphtheria toxoid, and acellular pertussis (Tdap) vaccines in pregnancy vaccine strain—a randomized, placebo-controlled study in twins. Vaccine.
in the Vaccine Adverse Event Reporting System (VAERS), 2011-2015. Vaccine. 2011;29:9508-9513.
2016;34:2349-2353. 560. Robinson CL, Advisory Committee on Immunization Practices ACAIWG. Advisory
490. Mortimer EA Jr. Pertussis and pertussis vaccine. In: Aronoff SC, ed. Advances Committee on Immunization Practices recommended immunization schedules
in Pediatric Infectious Diseases. Vol. 5. Chicago: Year Book Medical Publisher; for persons aged 0 through 18 years—United States, 2016. MMWR Morb Mortal
1990:1-27. Wkly Rep. 2016;65:86-87.
493. Murphy TV, Denniston MM, Hill HA, et al. Progress toward eliminating hepatitis 566. Routh J, Newton A, Mintz E. Cholera. In: Brunette G, ed. CDC Health Information
A disease in the United States. MMWR Suppl. 2016;65:29-41. for International Travel, 2016. New York: Oxford University Press; 2016.
495. Murphy TV. Haemophilus influenzae vaccines: 1997. Adv Pediatr Infect Dis. 568. Rubin LG, Levin MJ, Ljungman P, et al. 2013 IDSA clinical practice guideline for
1997;13:279-304. vaccination of the immunocompromised host. Clin Infect Dis. 2014;58:309-318.
497. National Institute of Allergy and Infectious Diseases. The Jordan Report—Accelerated 571. Safadi MA, Cintra OA. Epidemiology of meningococcal disease in Latin America:
Development of Vaccines, 2012. Washington, DC: National Institutes of Health; 2012. current situation and opportunities for prevention. Neurol Res. 2010;32:263-271.
500. Nelson GE, Aguon A, Valencia E, et al. Epidemiology of a mumps outbreak in a 575. Santolaya ME, O’Ryan M, Valenzuela MT, et al. Persistence of antibodies in adoles-
highly vaccinated island population and use of a third dose of measles-mumps- cents 18-24 months after immunization with one, two, or three doses of 4CMenB
rubella vaccine for outbreak control—Guam 2009 to 2010. Pediatr Infect Dis J. meningococcal serogroup B vaccine. Hum Vaccin Immunother. 2013;9:2304-2310.
2013;32:374-380. 576. Santolaya ME, O’Ryan ML, Valenzuela MT, et al. Immunogenicity and tolerability
503. Newton A, Routh J, Barbara E, Mahon B. Typhoid and paratyphoid fever. In: of a multicomponent meningococcal serogroup B (4CMenB) vaccine in healthy
Brunette G, ed. CDC Health Information for International Travel, 2016. New adolescents in Chile: a phase 2b/3 randomised, observer-blind, placebo-controlled
York: Oxford University Press; 2016. study. Lancet. 2012;379:617-624.
507. Nolan T, Richmond P, Marshall H, et al. Immunogenicity and safety of an 578. Sartorius B, Penttinen P, Nilsson J, et al. An outbreak of mumps in Sweden,
investigational combined Haemophilus influenzae type B-Neisseria meningiti- February-April 2004. Euro Surveill. 2005;10:191-193.
dis serogroups C and Y-tetanus toxoid conjugate vaccine. Pediatr Infect Dis J. 579. Satterwhite CL, Torrone E, Meites E, et al. Sexually transmitted infections among
2011;30:190-196. US women and men: prevalence and incidence estimates, 2008. Sex Transm Dis.
508. Noronha AS, Markowitz LE, Dunne EF. Systematic review of human papillomavirus 2013;40:187-193.
vaccine coadministration. Vaccine. 2014;32:2670-2674. 581. Schaffzin JK, Pollock L, Schulte C, et al. Effectiveness of previous mumps vac-
512. Onorato I, Markowitz L, Oxtoby M. Childhood immunization, vaccine-preventable cination during a summer camp outbreak. Pediatrics. 2007;120:e862-e868.
diseases and infection with human immunodeficiency virus. Pediatr Infect Dis 582. Schillie S, Murphy TV, Fenlon N, et al. Update: shortened interval for postvac-
J. 1988;6:588-595. cination serologic testing of infants born to hepatitis B-infected mothers. MMWR
516. Organization WH Global Polio Eradication Initiative. http://polioeradication.org/. Morb Mortal Wkly Rep. 2015;64:1118-1120.
520. Papania MJ, Wallace GS, Rota PA, et al. Elimination of endemic measles, rubella, 583. Schillie S, Walker T, Veselsky S, et al. Outcomes of infants born to women infected
and congenital rubella syndrome from the Western hemisphere: the US experience. with hepatitis B. Pediatrics. 2015;135:e1141-e1147.
JAMA Pediatr. 2014;168:148-155. 584. Schlegel M, Osterwalder JJ, Galeazzi RL, et al. Comparative efficacy of three
521. Parashar UD, Gibson CJ, Bresse JS, et al. Rotavirus and severe childhood diarrhea. mumps vaccines during disease outbreak in Eastern Switzerland: cohort study.
Emerg Infect Dis. 2006;12:304-306. BMJ. 1999;319:352.
527. Payne AB, Link-Gelles R, Azonobi I, et al. Invasive pneumococcal disease among 588. Schwarz AJ, Jackson JE, Ehrenkranz NJ, et al. Clinical evaluation of a new
children with and without sickle cell disease in the United States, 1998 to 2009. measles-mumps-rubella trivalent vaccine. Am J Dis Child. 1975;129:1408-1412.
Pediatr Infect Dis J. 2013;32:1308-1312. 591. Senders S, Bhuyan P, Jiang Q, et al. Immunogenicity, tolerability and safety
528. Payne DC, Edwards KM, Bowen MD, et al. Sibling transmission of vaccine- in adolescents of bivalent rLP2086, a meningococcal serogroup B vaccine,
derived rotavirus (RotaTeq) associated with rotavirus gastroenteritis. Pediatrics. coadministered with quadrivalent human papilloma virus vaccine. Pediatr Infect
2010;125:e438-e441. Dis J. 2016;35:548-554.
532. Peter G. Vaccine crisis: an emerging societal problem. J Infect Dis. 1985;151:981-983. 597. Sheldon EA, Schwartz H, Jiang Q, et al. A phase 1, randomized, open-label,
533. Petrosky E, Bocchini JA Jr, Hariri S, et al. Use of 9-valent human papillomavirus active-controlled trial to assess the safety of a meningococcal serogroup B bivalent
(HPV) vaccine: updated HPV vaccination recommendations of the advisory rLP2086 vaccine in healthy adults. Hum Vaccin Immunother. 2012;8:888-895.
committee on immunization practices. MMWR Morb Mortal Wkly Rep. 601. Shui IM, Baggs J, Patel M, et al. Risk of intussusception following administration
2015;64:300-304. of a pentavalent rotavirus vaccine in US infants. JAMA. 2012;307:598-604.
534. Phadke VK, Bednarczyk RA, Salmon DA, et al. Association between vaccine 607. Singh R, John TJ, Cherian T, et al. Immune response to measles, mumps &
refusal and vaccine-preventable diseases in the United States: a review of measles rubella vaccine at 9, 12 & 15 months of age. Indian J Med Res. 1994;100:155-159.
and pertussis. JAMA. 2016;315:1149-1158. 608. Singleton R, Hammitt L, Hennessy T, et al. The Alaska Haemophilus influenzae
540. Plotkin SA, Buser F. History of RA27/3 rubella vaccine. Rev Infect Dis. 1985;7(suppl type b experience: lessons in controlling a vaccine-preventable disease. Pediatrics.
1):S77-S78. 2006;118:e421-e429.
541. Plotkin SA. Increasing Complexity of Vaccine Development. J Infect Dis. 610. Smith CK, McNeal MM, Meyer NR, et al. Rotavirus shedding in premature
2015;212(suppl 1):S12-S16. infants following first immunization. Vaccine. 2011;29:8141-8146.
544. Popow-Kraupp T, Kundi M, Ambrosch F, et al. A controlled trial for evaluating 611. Smith PJ, Marcuse EK, Seward JF, et al. Children and adolescents unvaccinated
two live attenuated mumps-measles vaccines (Urabe Am 9-Schwarz and Jeryl against measles: geographic clustering, parents’ beliefs, and missed opportunities.
Lynn-Moraten) in young children. J Med Virol. 1986;18:69-79. Public Health Rep. 2015;130:485-504.
545. Preblud SR. Some current issues relating to rubella vaccine. JAMA. 612. Snijders BE, van Lier A, van de Kassteele J, et al. Mumps vaccine effective-
1985;254:253-256. ness in primary schools and households, the Netherlands, 2008. Vaccine.
546. Read RC, Baxter D, Chadwick DR, et al. Effect of a quadrivalent meningococcal 2012;30:2999-3002.
ACWY glycoconjugate or a serogroup B meningococcal vaccine on meningo- 614. Sotir MJ, Esposito DH, Barnett ED, et al. Measles in the 21st century, a continuing
coccal carriage: an observer-blind, phase 3 randomised clinical trial. Lancet. preventable risk to travelers: data from the GeoSentinel Global Network. Clin
2014;384:2123-2131. Infect Dis. 2016;62:210-212.
547. Read TR, Hocking JS, Chen MY, et al. The near disappearance of genital warts 617. Staples JE, Bocchini JA Jr, Rubin L, et al. Yellow fever vaccine booster doses:
in young women 4 years after commencing a national human papillomavirus recommendations of the Advisory Committee on Immunization Practices, 2015.
(HPV) vaccination programme. Sex Transm Infect. 2011;87:544-547. MMWR Morb Mortal Wkly Rep. 2015;64:647-650.
548. Redd SC, King GE, Heath JL, et al. Comparison of vaccination with measles- 620. Stokley S, Jeyarajah J, Yankey D, et al. Human papillomavirus vaccination coverage
mumps-rubella vaccine at 9, 12, and 15 months of age. J Infect Dis. 2004;189(suppl among adolescents, 2007-2013, and postlicensure vaccine safety monitoring,
1):S116-S122. 2006-2014—United States. MMWR Morb Mortal Wkly Rep. 2014;63:620-624.
549. Reef SE, Cochi SL. The evidence for the elimination of rubella and congenital 622. Stratton KR, Howe CJ, Johnston RB Jr. Adverse events associated with childhood
rubella syndrome in the United States: a public health achievement. Clin Infect vaccines other than pertussis and rubella. Summary of a report from the Institute
Dis. 2006;43(suppl 3):S123-S125. of Medicine. JAMA. 1994;271:1602-1605.
624. Sugg WC, Finger JA, Levine RH, et al. Field evaluation of live virus mumps 679. Weintraub ES, Baggs J, Duffy J, et al. Risk of Intussusception after monovalent
vaccine. J Pediatr. 1968;72:461-466. rotavirus vaccination. N Engl J Med. 2014;370:513-519.
625. Sukumaran L, McCarthy NL, Kharbanda EO, et al. Association of Tdap vaccination 684. Wharton M, Cochi SL, Hutcheson RH, et al. A large outbreak of mumps in the
with acute events and adverse birth outcomes among pregnant women with postvaccine era. J Infect Dis. 1988;158:1253-1260.
prior tetanus-containing immunizations. JAMA. 2015;314:1581-1587. 687. Giersing BK, Modjarrad K, Kaslow DC, Moorthy VS. Report from the World
627. Sun Y, Christensen J, Hviid A, et al. Risk of febrile seizures and epilepsy after Health Organization’s Product Development for Vaccines Advisory Committee
vaccination with diphtheria, tetanus, acellular pertussis, inactivated poliovirus, (PDVAC) meeting, Geneva, September 7-9, 2015. WHO Product Development
and Haemophilus influenzae type B. JAMA. 2012;307:823-831. for Vaccines Advisory Committee; WHO Product Development for Vaccines
632. Tate JE, Mutuc JD, Panozzo CA, et al. Sustained decline in rotavirus detections Product Development Advisory Committee. Vaccine. 2016;34:2865-2869.
in the United States following the introduction of rotavirus vaccine in 2006. 688. Widdowson MA, Meltzer MI, Zhang X, et al. Cost-effectiveness and potential
Pediatr Infect Dis J. 2011;30(suppl):S30-S34. impact of rotavirus vaccination in the United States. Pediatrics. 2007;119:684-697.
639. Tischer A, Gerike E. Immune response after primary and re-vaccination with different 689. Willis E, Marko A, Marin M, et al. 1048Pregnancy registry for varicella-zoster
combined vaccines against measles, mumps, rubella. Vaccine. 2000;18:1382-1392. virus-containing vaccines: 18-year summary of pregnancy outcomes. Open Forum
640. Toneatto D, Ismaili S, Ypma E, et al. The first use of an investigational multi- Infect Dis. 2014;1(suppl 1):S307.
component meningococcal serogroup B vaccine (4CMenB) in humans. Hum 691. Winter K, Glaser C, Watt J, et al. Pertussis epidemic—California, 2014. MMWR
Vaccin. 2011;7:646-653. Morb Mortal Wkly Rep. 2014;63:1129-1132.
641. Toscani L, Batou M, Bouvier P, et al. Comparison of the efficacy of various 694. Witte JJ, Karchmer AW, Case G, et al. Epidemiology of rubella. Am J Dis Child.
strains of mumps vaccine: a school survey. Soz Praventivmed. 1996;41:341-347. 1969;118:107-111.
647. Usonis V, Bakasenas V, Chitour K, et al. Comparative study of reactogenicity and 696. World Health Organization. Global Advisory Committee on Vaccine Safety, report
immunogenicity of new and established measles, mumps and rubella vaccines of meeting held 3-4 December 2009. Wkly Epidemiol Rec. 2010;85:29-33.
in healthy children. Infection. 1998;26:222-226. 697. World Health Organization. Haemophilus influenzae type b (Hib) vaccination
648. Usonis V, Bakasenas V, Kaufhold A, et al. Reactogenicity and immunogenicity of position paper—July 2013. Wkly Epidemiol Rec. 2013;88:413-426.
a new live attenuated combined measles, mumps and rubella vaccine in healthy 698. World Health Organization. Japanese encephalitis vaccines: WHO position
children. Pediatr Infect Dis J. 1999;18:42-48. paper—February 2015. Wkly Epidemiol Rec. 2015;90:69-87.
653. Vandermeulen C, Clement F, Roelants M, et al. Evaluation of cellular immunity 699. World Health Organization. Polio vaccines: WHO position paper—March, 2016.
to mumps in vaccinated individuals with or without circulating antibodies up Wkly Epidemiol Rec. 2016;91:145-168.
to 16 years after their last vaccination. J Infect Dis. 2009;199:1457-1460. 701. World Health Organization. WHO position on measles vaccines. Vaccine.
665. Vu DM, Welsch JA, Zuno-Mitchell P, et al. Antibody persistence 3 years after 2009;27:7219-7221.
immunization of adolescents with quadrivalent meningococcal conjugate vaccine. 702. Wright JG, Quinn CP, Shadomy S, et al. Use of anthrax vaccine in the United
J Infect Dis. 2006;193:821-828. States: recommendations of the Advisory Committee on Immunization Practices
668. Walter EB, Neuzil KM, Zhu Y, et al. Influenza vaccine immunogenicity in 6- to (ACIP), 2009. MMWR Recomm Rep. 2010;59(RR-6):1-30.
23-month-old children: are identical antigens necessary for priming? Pediatrics. 705. Xie O, Pollard AJ, Mueller JE, et al. Emergence of serogroup X meningococcal
2006;118:e570-e578. disease in Africa: need for a vaccine. Vaccine. 2013;31:2852-2861.
669. Ward B, Boulinanne N, Ratnam S, et al. Cellular immunity in measles vaccine 706. Yang YT, Debold V. A longitudinal analysis of the effect of nonmedical exemption
failure: demonstration of measles antigen-specific lymphoproliferative responses law and vaccine uptake on vaccine-targeted disease rates. Am J Public Health.
despite limited serum antibody production after revaccination. J Infect Dis. 2014;104:371-377.
1995;172:1591-1595. 708. Yih WK, Lieu TA, Kulldorff M, et al. Intussusception risk after rotavirus vaccination
670. Wassilak SG, Oberste MS, Tangermann RH, et al. Progress toward global inter- in U.S. Infants. N Engl J Med. 2014;370:503-512.
ruption of wild poliovirus transmission, 2010-2013, and tackling the challenges 709. Yun K, Urban K, Mamo B, et al. Increasing hepatitis B vaccine prevalence among
to complete eradication. J Infect Dis. 2014;210(suppl 1):S5-S15. refugee children arriving in the United States, 2006-2012. Am J Public Health.
676. Weibel RE, Villarejos VM, Klein EB, et al. Clinical and laboratory studies of live 2016;106:1460-1462.
attenuated RA 27/3 and HPV 77-DE rubella virus vaccines. Proc Soc Exp Biol 716. Zimmerman RK, Nowalk MP, Chung J, et al. 2014-2015 Influenza vaccine
Med. 1980;165:44-49. effectiveness in the United States by vaccine type. Clin Infect Dis. 2016;63:
678. Weinberg MS. Vaccine recommendations for infants and children. In: Brunette 1564-1573.
G, ed. CDC Health Information for International Travel, 2016. New York: Oxford
University Press; 2016:490-521. The full reference list for this chapter is available at ExpertConsult.com.
CHAPTER 245  Active Immunizing Agents 2601.e1

24. American Academy of Pediatrics Committee on Infectious Diseases. Prevention


REFERENCES of rotavirus disease: updated guidelines for use of rotavirus vaccine. Pediatrics.
1. Abu Raya B, Srugo I, Kessel A, et al. The decline of pertussis-specific antibodies 2009;123:1412-1420.
after tetanus, diphtheria, and acellular pertussis immunization in late pregnancy. 25. American Academy of Pediatrics Committee on Infectious Diseases. Recom-
J Infect Dis. 2015;212:1869-1873. mendations for the prevention of Streptococcus pneumoniae infections in infants
2. Acharya IL, Lowe CU, Thapa R, et al. Prevention of typhoid fever in Nepal with and children: use of 13-valent pneumococcal conjugate vaccine (PCV13) and
the Vi capsular polysaccharide of Salmonella typhi. A preliminary report. N Engl pneumococcal polysaccharide vaccine (PPSV23). Pediatrics. 2010;126:186.
J Med. 1987;317:1101-1104. 26. American Academy of Pediatrics Committee on Infectious Diseases. Policy
3. Acosta CJ, Galindo CM, Ali M, et al. A multi-country cluster randomized controlled statement-Prevention of varicella: update of recommendations for use of quadri-
effectiveness evaluation to accelerate the introduction of Vi polysaccharide typhoid valent and monovalent varicella vaccines in children. Pediatrics. 2011;128:630-632.
vaccine in developing countries in Asia: rationale and design. Trop Med Int 27. American Academy of Pediatrics Committee on Infectious Diseases. Recom-
Health. 2005;10:1219-1228. mendations for administering hepatitis A vaccine to contacts of international
4. Adams DA, Thomas KR, Jajosky RA, et al. Summary of notifiable infectious adoptees. Pediatrics. 2011;128:803-804.
diseases and conditions—United States, 2014. MMWR Morb Mortal Wkly Rep. 28. American Academy of Pediatrics Committee on Infectious Diseases. Updated rec-
2016;63:1-152. ommendations on the use of meningococcal vaccines. Pediatrics. 2014;134:400-403.
5. Advisory Committee on Immunization P, Centers for Disease Control and 29. American Academy of Pediatrics. Active immunization. In: Kimberlin DW,
Prevention. Immunization of health-care personnel: recommendations of the Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee
Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep. on Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of
2011;60(RR-7):1-45. Pediatrics; 2015:13-43.
6. Ag Ayoya M, Higgins-Steele A, Massai D, et al. Health authorities’ leadership 30. American Academy of Pediatrics. Arboviruses. In: Kimberlin DW, Brady MT,
reduces cholera deaths in Haiti. Lancet. 2012;380:473-474. Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infec-
7. Albertson JP, Clegg WJ, Reid HD, et al. Mumps outbreak at a university and tious Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics;
recommendation for a third dose of measles-mumps-rubella vaccine—Illinois, 2015:240-246.
2015-2016. MMWR Morb Mortal Wkly Rep. 2016;65:731-734. 31. American Academy of Pediatrics. Diphtheria. In: Kimberlin DW, Brady MT, Jackson
8. Ali M, Nelson AR, Lopez AL, et al. Updated global burden of cholera in endemic MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious Diseases.
countries. PLoS Negl Trop Dis. 2015;9:e0003832. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:325-329.
9. Aliabadi N, Tate JE, Haynes AK, et al. Sustained decrease in laboratory detection 32. American Academy of Pediatrics. Haemophilus influenzae infections. In: Kimberlin
of rotavirus after implementation of routine vaccination-United States, 2000-2014. DW, Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee
MMWR Morb Mortal Wkly Rep. 2015;64:337-342. on Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of
10. Allison MA, Daley MF, Crane LA, et al. Influenza vaccine effectiveness in Pediatrics; 2015:368-376.
healthy 6- to 21-month-old children during the 2003-2004 season. J Pediatr. 33. American Academy of Pediatrics. Hepatitis A. In: Kimberlin DW, Brady MT, Jackson
2006;149:755-762. MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious Diseases.
11. Alter MJ, Hadler SC, Margolis HS, et al. The changing epidemiology of hepa- 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:391-399.
titis B in the United States. Need for alternative vaccination strategies. JAMA. 34. American Academy of Pediatrics. Hepatitis B. In: Kimberlin DW, Brady MT, Jackson
1990;263:1218-1222. MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious Diseases.
12. Ambrosch F, Wiedermann G, Crooy P, et al. Immunogenicity and side-effects 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:400-423.
of a new tetravalent meningococcal polysaccharide vaccine. Bull World Health 35. American Academy of Pediatrics. Human papilloma viruses. In: Kimberlin DW,
Organ. 1983;61:317-323. Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee
13. American Academy of Pediatrics Committee on Infectious Diseases. Measles: on Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of
reassessment of the current immunization policy. Pediatrics. 1989;84: Pediatrics; 2015:576-583.
1110-1113. 36. American Academy of Pediatrics. Immunization in special clinical circumstances.
14. American Academy of Pediatrics Committee on Infectious Diseases. The relation- In: Kimberlin DW, Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report
ship between pertussis vaccine and central nervous system sequelae: continuing of the Committee on Infectious Diseases. 30th ed. Elk Grove Village, IL: American
assessment. American Academy of Pediatrics Committee on Infectious Diseases. Academy of Pediatrics; 2015:68-107.
Pediatrics. 1996;97:279-281. 37. American Academy of Pediatrics. Influenza. In: Kimberlin DW, Brady MT, Jackson
15. American Academy of Pediatrics Committee on Infectious Diseases. Immunization MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious Diseases.
of adolescents: recommendations of the Advisory Committee on Immunization 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:476-493.
Practices, the American Academy of Pediatrics, the American Academy of Family 38. American Academy of Pediatrics. Measles. In: Kimberlin DW, Brady MT, Jackson
Physicians, and the American Medical Association. American Academy of Pediatrics MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious Diseases.
Committee on Infectious Diseases. Pediatrics. 1997;99:479-488. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:535-547.
16. American Academy of Pediatrics Committee on Infectious Diseases. Poliomyelitis 39. American Academy of Pediatrics. Meningococcal infections. In: Kimberlin DW,
prevention: recommendations for use of inactivated poliovirus vaccine and live Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee
oral poliovirus vaccine. American Academy of Pediatrics Committee on Infectious on Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of
Diseases. Pediatrics. 1997;99:300-305. Pediatrics; 2015:547-558.
17. American Academy of Pediatrics Committee on Infectious Diseases and Committee 40. American Academy of Pediatrics. Mumps. In: Kimberlin DW, Brady MT, Jackson
on Pediatric AIDS. Measles immunization in HIV-infected children. American MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious Diseases.
Academy of Pediatrics. Committee on Infectious Diseases and Committee on 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:564-568.
Pediatric AIDS. Pediatrics. 1999;103:1057-1060. 41. American Academy of Pediatrics. Pertussis (whooping cough). In: Kimberlin DW,
18. American Academy of Pediatrics Committee on Infectious Diseases. Poliomyelitis Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee
prevention: revised recommendations for use of inactivated and live oral poliovirus on Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of
vaccines. American Academy of Pediatrics Committee on Infectious Diseases. Pediatrics; 2015:608-621.
Pediatrics. 1999;103:171-172. 42. American Academy of Pediatrics. Pneumococcal infections. In: Kimberlin DW,
19. American Academy of Pediatrics Committee on Infectious Diseases. Prevention Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee
of poliomyelitis: recommendations for use of only inactivated poliovirus vaccine on Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of
for routine immunization. Pediatrics. 1999;104:1404-1406. Pediatrics; 2015:626-638.
20. American Academy of Pediatrics. Committee on Native American Child Health 43. American Academy of Pediatrics. Poliovirus infections. In: Kimberlin DW,
and Committee on Infectious Diseases. Immunizations for Native American Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee
children. American Academy of Pediatrics. Pediatrics. 1999;104:564-567. on Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of
21. American Academy of Pediatrics Committee on Infectious Diseases. Prevention Pediatrics; 2015:644-650.
of pertussis among adolescents: recommendations for use of tetanus toxoid, 44. American Academy of Pediatrics. Rubella. In: Kimberlin DW, Brady MT, Jackson
reduced diphtheria toxoid, and acellular pertussis (Tdap) vaccine. Pediatrics. MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infectious Diseases.
2006;117:965-978. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2015:688-695.
22. American Academy of Pediatrics Committee on Infectious Diseases. Hepatitis 45. American Academy of Pediatrics. Salmonella infections. In: Kimberlin DW,
A vaccine recommendations. Pediatrics. 2007;120:189-199. Brady MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee
23. American Academy of Pediatrics Committee on Infectious Diseases. Prevention of on Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of
varicella: recommendations for use of varicella vaccines in children, including a Pediatrics; 2015:695-702.
recommendation for a routine 2-dose varicella immunization schedule. Pediatrics. 46. American Academy of Pediatrics. Smallpox (variola). In: Kimberlin DW, Brady
2007;120:221-231. MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on
2601.e2 PART V  Prevention of Infectious Diseases

Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 75. Bernstein HH, Rothstein EP, Watson BM, et al. Clinical survey of natural varicella
2015:709-712. compared with breakthrough varicella after immunization with live attenuated
47. American Academy of Pediatrics. Tetanus (lockjaw). In: Kimberlin DW, Brady Oka/Merck varicella vaccine. Pediatrics. 1993;92:833-837.
MT, Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on 76. Bernstein HH, Starke JR, American Academy of Pediatrics. Committee on Infec-
Infectious Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; tious Diseases. Policy statement—recommendation for mandatory influenza
2015:773-778. immunization of all health care personnel. Pediatrics. 2010;126:809-815.
48. American Academy of Pediatrics. Tuberculosis. In: Kimberlin DW, Brady MT, 77. Bewley KM, Schwab JG, Ballanco GA, et al. Interchangeability of Haemophilus
Jackson MA, Long SS, eds. Red Book: 2015 Report of the Committee on Infec- influenzae type b vaccines in the primary series: evaluation of a two-dose mixed
tious Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; regimen. Pediatrics. 1996;98:898-904.
2015:805-831. 78. Bilukha OO, Rosenstein N, National Center for Infectious Diseases, et al.
49. American Academy of Pediatrics. Varicella-zoster. In: Kimberlin DW, Brady MT, Prevention and control of meningococcal disease. Recommendations of the
Jackson MA, Long SS, eds. Red Book: 2012 Report of the Committee on Infec- Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep.
tious Diseases. 30th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2005;54(RR-7):1-21.
2015:846-860. 79. Black S, Greenberg DP. A combined diphtheria, tetanus, five-component acellular
50. Amirthalingam G, Andrews N, Campbell H, et al. Effectiveness of maternal pertus- pertussis, poliovirus and Haemophilus influenzae type b vaccine. Expert Rev
sis vaccination in England: an observational study. Lancet. 2014;384:1521-1528. Vaccines. 2005;4:793-805.
51. Ammann AJ, Addiego J, Wara DW, et al. Polyvalent pneumococcal-polysaccharide 80. Black S, Shinefield H, Fireman B, et al. Efficacy, safety and immunogenicity of hep-
immunization of patients with sickle-cell anemia and patients with splenectomy. tavalent pneumococcal conjugate vaccine in children. Northern California Kaiser
N Engl J Med. 1977;297:897-900. Permanente Vaccine Study Center Group. Pediatr Infect Dis J. 2000;19:187-195.
52. Ampofo K. The changing epidemiology of invasive pneumococcal disease at a 81. Black CL, Yue X, Ball SW, et al. Influenza Vaccination Coverage Among Health
tertiary children’s hospital through the 7-valent pneumococcal conjugate vaccine Care Personnel—United States, 2014-15 Influenza Season. MMWR Morb Mortal
era: a case for continuous surveillance. Pediatr Infect Dis J. 2012;31:228. Wkly Rep. 2015;64:993-999.
53. Anderson EL, Decker MD, Englund JA, et al. Interchangeability of conjugated 82. Bloch AB, Orenstein WA, Stetler HC, et al. Health impact of measles vaccination
Haemophilus influenzae type b vaccines in infants. JAMA. 1995;273:849-853. in the United States. Pediatrics. 1985;76:524-532.
54. Anh DD. Safety and immunogenicity of a reformulated Vietnamese bivalent 83. Block SL, Vesikari T, Goveia MG, et al. Efficacy, immunogenicity, and safety of
killed, whole-cell, oral cholera vaccine in adults. Vaccine. 2007;25:1149. a pentavalent human-bovine (WC3) reassortant rotavirus vaccine at the end of
55. Arbeter AM, Starr SE, Plotkin SA. Varicella vaccine studies in healthy children shelf life. Pediatrics. 2007;119:11-18.
and adults. Pediatrics. 1986;78(suppl):748-756. 84. Booy R, Moxon ER, MacFarlane JA, et al. Efficacy of Haemophilus influenzae
56. Arguedas A, Soley C, Lindert K. Responses to 2009 H1N1 vaccine in children type B conjugate vaccine in Oxford region. Lancet. 1992;340:847.
3 to 17 years of age. N Engl J Med. 2010;362:370-372. 85. Borgono JM, Greiber R, Solari G, et al. A field trial of combined measles-mumps-
57. Armand J, Arminjon F, Mynard MC, et al. Tetravalent meningococcal polysac- rubella vaccine. Satisfactory immunization with 188 children in Chile. Clin Pediatr
charide vaccine groups A, C, Y, W 135: clinical and serological evaluation. J Biol (Phila). 1973;12:170-2.
Stand. 1982;10:335-339. 86. Bradley JS, Peacock G, Krug SE, et al. Pediatric anthrax clinical management.
58. Arpadi SM, Markowitz LE, Baughman AL, et al. Measles antibody in vac- Pediatrics. 2014;133:e1411-e1436.
cinated human immunodeficiency virus type 1-infected children. Pediatrics. 87. Brady MT, Byington CL, Davies HD, et al. Immunization for Streptococcus
1996;97:653-657. pneumoniae infections in high-risk children. Pediatrics. 2014;134:1230-1233.
59. Asano Y, Nagai T, Miyata T, et al. Long-term protective immunity of recipients 88. Breiman RF, Butler JC, Tenover FC, et al. Emergence of drug-resistant pneumococ-
of the OKA strain of live varicella vaccine. Pediatrics. 1985;75:667-671. cal infections in the United States. JAMA. 1994;271:1831-1835.
60. Atwell JE, Van Otterloo J, Zipprich J, et al. Nonmedical vaccine exemptions and 89. Briere EC, Rubin L, Moro PL, et al. Prevention and control of Haemophilus
pertussis in California, 2010. Pediatrics. 2013;132:624-630. influenzae type b disease: recommendations of the advisory committee on
61. Bakare N, Menschik D, Tiernan R, et al. Severe combined immunodeficiency immunization practices (ACIP). MMWR Recomm Rep. 2014;63(RR-01):1-14.
(SCID) and rotavirus vaccination: reports to the Vaccine Adverse Events Reporting 90. Broder KR, Cortese MM, Iskander JK, et al. Preventing tetanus, diphtheria,
System (VAERS). Vaccine. 2010;28:6609-6612. and pertussis among adolescents: use of tetanus toxoid, reduced diphtheria
62. Ball R, Braun MM, Mootrey GT, et al. Safety data on meningococcal polysac- toxoid and acellular pertussis vaccines recommendations of the Advisory
charide vaccine from the Vaccine Adverse Event Reporting System. Clin Infect Committee on Immunization Practices (ACIP). MMWR Recomm Rep.
Dis. 2001;32:1273-1280. 2006;55(RR-3):1-34.
63. Barbour ML. Conjugate vaccines and the carriage of Haemophilus influenzae 91. Brokstad KA, Cox RJ, Olofsson J, et al. Parenteral influenza vaccination induces
type b. Emerg Infect Dis. 1996;2:176-182. a rapid systemic and local immune response. J Infect Dis. 1995;171:198-203.
64. Barker WH. Excess pneumonia and influenza associated hospitalization 92. Broliden K, Abreu ER, Arneborn M, et al. Immunity to mumps before and after
during influenza epidemics in the United States, 1970-78. Am J Public Health. MMR vaccination at 12 years of age in the first generation offered the two-dose
1986;76:761-765. immunization programme. Vaccine. 1998;16:323-327.
65. Barker WH, Mullooly JP. Impact of epidemic type A influenza in a defined adult 93. Brooks R, Woods CW, Benjamin DK Jr, et al. Increased case-fatality rate
population. Am J Epidemiol. 1980;112:798-811. associated with outbreaks of Neisseria meningitidis infection, compared with
66. Barlow WE, Davis RL, Glasser JW, et al. The risk of seizures after receipt of sporadic meningococcal disease, in the United States, 1994-2002. Clin Infect
whole-cell pertussis or measles, mumps, and rubella vaccine. N Engl J Med. Dis. 2006;43:49-54.
2001;345:656-661. 94. Brunell PA, Argaw T. Chickenpox attributable to a vaccine virus contracted from
67. Barskey AE, Glasser JW, LeBaron CW. Mumps resurgences in the United a vaccinee with zoster. Pediatrics. 2000;106:E28.
States: a historical perspective on unexpected elements. Vaccine. 2009;27: 95. Bryan J, Henry C, Hoffman A, et al. Randomized, cross-over, controlled comparison
6186-6195. of two inactivated hepatitis A vaccines. Vaccine. 2000;19:743-750.
68. Baylis SA, Finsterbusch T, Bannert N, et al. Analysis of porcine circovirus type 96. Bryant KA, Marshall GS, Marchant CD, et al. Immunogenicity and safety of H
1 detected in Rotarix vaccine. Vaccine. 2011;29:690-697. influenzae type b-N meningitidis C/Y conjugate vaccine in infants. Pediatrics.
69. Beasley RP, Detels R, Kim KS, et al. Prevention of rubella during an epidemic 2011;127:e1375-e1385.
on Taiwan. HPV-77 and RA 27-3 rubella vaccines administered subcutaneously 97. Buckle GC, Walker CL, Black RE. Typhoid fever and paratyphoid fever: systematic
and intranasally HPV-77 vaccine mixed with mumps and-or measles vaccines. review to estimate global morbidity and mortality for 2010. J Glob Health.
Am J Dis Child. 1969;118:301-306. 2012;2:010401.
70. Beeler J, Varricchio F, Wise R. Thrombocytopenia after immunization with measles 98. Burns CC, Diop OM, Sutter RW, et al. Vaccine-derived polioviruses. J Infect Dis.
vaccines: review of the vaccine adverse events reporting system (1990 to 1994). 2014;210(suppl 1):S283-S293.
Pediatr Infect Dis J. 1996;15:88-90. 99. Butler JC, Breiman RF, Campbell JF, et al. Pneumococcal polysaccharide vaccine
71. Bell TD, Chai H, Berlow B, et al. Immunization with killed influenza virus in efficacy. An evaluation of current recommendations. JAMA. 1993;270:1826-1831.
children with chronic asthma. Chest. 1978;73:140-145. 100. Buttery JP, Danchin MH, Lee KJ, et al. Intussusception following rotavirus vaccine
72. Bell BP, Negus S, Fiore AE, et al. Immunogenicity of an inactivated hepatitis A administration: post-marketing surveillance in the National Immunization
vaccine in infants and young children. Pediatr Infect Dis J. 2007;26:116-122. Program in Australia. Vaccine. 2011;29:3061-3066.
73. Belongia EA, Irving SA, Shui IM, et al. Real-time surveillance to assess risk 101. Byington CL, Maldonado YA, Barnett ED, et al. Recommendations for preven-
of intussusception and other adverse events after pentavalent, bovine-derived tion and control of influenza in children, 2016–2017. Pediatrics. 2016;138:pii:
rotavirus vaccine. Pediatr Infect Dis J. 2010;29:1-5. e20162527.
74. Belshe R, Walker RI, Stoddard J, et al. Influenza vaccine—live. In: Plotkin SA, 102. Byington CL, Samore MH, Stoddard GJ, et al. Temporal trends of invasive disease
Orenstein WA, offit PA, eds. Vaccines. 5th ed. Philadelphia: Saunders Elsevier; due to Streptococcus pneumoniae among children in the intermountain west:
2008:291-309. emergence of nonvaccine serogroups. Clin Infect Dis. 2005;41:21-29.
CHAPTER 245  Active Immunizing Agents 2601.e3

103. Caceres VM, Sutter RW. Sabin monovalent oral polio vaccines: review of past 130. Centers for Disease Control and Prevention. Prevention of pneumococcal disease:
experiences and their potential use after polio eradication. Clin Infect Dis. Recommendations of the Advisory Committee on Immunization Practices (ACIP).
2001;33:531-541. MMWR Recomm Rep. 1997;46(RR-8):1-24.
104. Castilla J, Garcia Cenoz M, Arriazu M, et al. Effectiveness of Jeryl Lynn-containing 131. Centers for Disease Control and Prevention. Impact of the sequential IPV/OPV
vaccine in Spanish children. Vaccine. 2009;27:2089-2093. schedule on vaccination coverage levels—United States, 1997. MMWR Morb
105. Centers for Disease Control. Poliomyelitis prevention: recommendations of the Mortal Wkly Rep. 1998;47:1017-1019.
Immunization Practices Advisory Committee (ACIP). MMWR Morb Mortal 132. Centers for Disease Control and Prevention. Human rabies prevention—United
Wkly Rep. 1982;31:22-26, 31-34. States, 1999. Recommendations of the Advisory Committee on Immunization
106. Centers for Disease Control. Subacute sclerosing panencephalitis surveillance— Practices (ACIP). MMWR Morb Mortal Wkly Rep. 1999;48(RR-1):1-21.
United States. MMWR Morb Mortal Wkly Rep. 1982;31:585-588. 133. Centers for Disease Control and Prevention. Prevention of varicella: updated
107. Centers for Disease Control. Rubella and congenital rubella—United States, recommendations of the Advisory Committee on Immunization Practices (ACIP).
1983. MMWR Morb Mortal Wkly Rep. 1984;33:237-242, 247. MMWR Recomm Rep. 1999;48(RR-6):1-5.
108. Centers for Disease Control. Systemic allergic reactions following immuniza- 134. Centers for Disease Control and Prevention. Recommendations of the Advisory
tion with human diploid cell rabies vaccine. MMWR Morb Mortal Wkly Rep. Committee on Immunization Practices: revised recommendations for routine
1984;33:185-187. poliomyelitis vaccination. MMWR Morb Mortal Wkly Rep. 1999;48:590.
109. Centers for Disease Control. Measles prevention. MMWR Suppl. 1989;38:1-18. 135. Centers for Disease Control and Prevention. Update: recommendations to prevent
110. Centers for Disease Control. Mumps prevention. MMWR Morb Mortal Wkly hepatitis B virus transmission—United States. MMWR Morb Mortal Wkly Rep.
Rep. 1989;38:388-392, 397-400. 1999;48:33-34.
111. Centers for Disease Control. Mumps—United States, 1985-1988. MMWR Morb 136. Centers for Disease Control and Prevention. Notice to readers: alternate two-dose
Mortal Wkly Rep. 1989;38:101-105. hepatitis B vaccination schedule for adolescents aged 11–15 years. MMWR Morb
112. Centers for Disease Control. Pneumococcal polysaccharide vaccine: recommenda- Mortal Wkly Rep. 2000;49:261.
tions of the Advisory Committee on Immunization Practices (ACIP). MMWR. 137. Centers for Disease Control and Prevention. Notice to readers: recommended
1989;38:64-68, 73-76. childhood immunization schedule—United States, 2000. MMWR Morb Mortal
113. Centers for Disease Control. Rubella vaccination during pregnancy—United Wkly Rep. 2000;49:35-38, 47.
States, 1971-1988. MMWR Morb Mortal Wkly Rep. 1989;38:289-293. 138. Centers for Disease Control and Prevention. Use of diphtheria toxoid-tetanus
114. Centers for Disease Control. Surveillance of tetanus, 1987 and 1988. Wkly Epidemiol toxoid-acellular pertussis vaccine as a five-dose series: supplemental recom-
Rec. 1990;65:261-263. mendations of the Advisory Committee on Immunization Practices (ACIP).
115. Centers for Disease Control. Diphtheria, tetanus, and pertussis: recommenda- MMWR Morb Mortal Wkly Rep. 2000;49(RR-13).
tions for vaccine use and other preventive measures: recommendations of the 139. Centers for Disease Control and Prevention. Revised ACIP recommendation for
Immunization Practices Advisory Committee (ACIP). MMWR Morb Mortal avoiding pregnancy after receiving a rubella-containing vaccine. MMWR Morb
Wkly Rep. 1991;40(RR-10):1-28. Mortal Wkly Rep. 2001;50:1117.
116. Centers for Disease Control. Hepatitis B virus: a comprehensive strategy for 140. Centers for Disease Control and Prevention. Adverse events associated with
eliminating transmission in the United States through universal childhood vac- 17D-derived yellow fever vaccination—United States, 2001-2002. MMWR Morb
cination. Recommendations of the Immunization Practices Advisory Committee Mortal Wkly Rep. 2002;51:989-993.
(ACIP). MMWR Recomm Rep. 1991;40(RR-13):1-25. 141. Centers for Disease Control and Prevention. General recommendations on
117. Centers for Disease Control. Rabies prevention—United States, 1991. Recom- immunization. Recommendations of the Advisory Committee on Immunization
mendations of the Immunization Practices Advisory Committee (ACIP). MMWR Practices (ACIP) and the American Academy of Family Physicians (AAFP).
Morb Mortal Wkly Rep. 1991;40(RR-3):1-19. MMWR. 2002;51(RR-2):1-35.
118. Centers for Disease Control and Prevention. Rabies prevention—United States, 142. Centers for Disease Control and Prevention. Pertussis—United States, 1997-2000.
1984. MMWR Morb Mortal Wkly Rep. 1984;33:393-402, 407-408. MMWR Morb Mortal Wkly Rep. 2002;51:73-76.
119. Deleted in review. 143. Centers for Disease Control and Prevention. Progress toward elimination of
120. Centers for Disease Control and Prevention. Progress towards the global Haemophilus influenzae type b invasive disease among infants and children—
elimination of neonatal tetanus, 1989-1993. MMWR Morb Mortal Wkly Rep. United States, 1998-2000. MMWR Morb Mortal Wkly Rep. 2002;51:234-237.
1994;43:885-887, 893-894. 144. Centers for Disease Control and Prevention. Pneumococcal vaccination for
121. Centers for Disease Control and Prevention. Rubella and congenital rubella cochlear implant candidates and recipients: updated recommendations of the
syndrome—United States, January 1, 1991-May 7, 1994. MMWR Morb Mortal Advisory Committee on Immunization Practices. MMWR Morb Mortal Wkly
Wkly Rep. 1994;43:391, 397-401. Rep. 2003;52:739-740.
122. Centers for Disease Control and Prevention. Cholera associated with food 145. Centers for Disease Control and Prevention. Direct and indirect effects of routine
transported from El Salvador—Indiana, 1994. MMWR Morb Mortal Wkly Rep. vaccination of children with 7-valent pneumococcal conjugate vaccine on incidence
1995;44:385-386. of invasive pneumococcal disease—United States, 1998-2003. MMWR Morb
123. Centers for Disease Control and Prevention. Pertussis—United States, January Mortal Wkly Rep. 2005;54:893-897.
1992-June 1995. MMWR Morb Mortal Wkly Rep. 1995;44:525-529. 146. Centers for Disease Control and Prevention. Elimination of rubella and congenital
124. Centers for Disease Control and Prevention. Update: recommendations to prevent rubella syndrome—United States, 1969-2004. MMWR Morb Mortal Wkly Rep.
hepatitis B virus transmission—United States. MMWR Morb Mortal Wkly Rep. 2005;54:279-282.
1995;44:574-575. 147. Centers for Disease Control and Prevention. National, state, and urban area
125. Centers for Disease Control and Prevention. Immunization of adolescents: vaccination coverage among children aged 19-35 months—United States, 2004.
recommendations of the Advisory Committee on Immunization Practices, MMWR Morb Mortal Wkly Rep. 2005;54:717-721.
the American Academy of Pediatrics, the American Academy of Family Physi- 148. Centers for Disease Control and Prevention. Notice to readers: licensure of a
cians and the American Medical Association. MMWR Morb Mortal Wkly Rep. combined measles, mumps, rubella and varicella vaccine. MMWR Morb Mortal
1996;45(RR-13):1-16. Wkly Rep. 2005;54:1212.
126. Centers for Disease Control and Prevention. Measles pneumonitis following 149. Centers for Disease Control and Prevention. Inadvertent misadministration of
measles-mumps-rubella vaccination of a patient with HIV infection, 1993. meningococcal conjugate vaccine—United States, June-August 2005. MMWR
MMWR Morb Mortal Wkly Rep. 1996;45:603-606. Morb Mortal Wkly Rep. 2006;55:1016-1017.
127. Centers for Disease Control and Prevention. The role of BCG vaccine in the 150. Centers for Disease Control and Prevention. Notice to readers: updated recom-
prevention and control of tuberculosis in the United States: a joint statement by mendations of the Advisory Committee on Immunization Practices (ACIP)
the Advisory Council for the Elimination of Tuberculosis and the Advisory for the control and elimination of mumps. MMWR Morb Mortal Wkly Rep.
Committee on Immunization Practices. MMWR Morb Mortal Wkly Rep. 1996; 2006;55:629-630.
45(RR-4):1-18. 151. Centers for Disease Control and Prevention. Update: prevention of hepatitis
128. Centers for Disease Control and Prevention. Update: vaccine side effects, adverse A after exposure to hepatitis A virus and in international travelers. Updated
reactions, contraindications, and precautions. Recommendations of the Advisory recommendations of the Advisory Committee on Immunization Practices (ACIP).
Committee on Immunization Practices (ACIP) [published erratum appears in MMWR Morb Mortal Wkly Rep. 2007;56:1080-1084.
MMWR Morb Mortal Wkly Rep 1997 Mar 14;46:227]. MMWR Morb Mortal 152. Centers for Disease Control and Prevention. Licensure of a diphtheria and tetanus
Wkly Rep. 1996;45(RR-12):1-35. toxoids and acellular pertussis adsorbed, inactivated poliovirus, and Haemophilus
129. Centers for Disease Control and Prevention. Poliomyelitis prevention in the B conjugate vaccine and guidance for use in infants and children. MMWR Morb
United States: introduction of a sequential vaccination schedule of inactivated Mortal Wkly Rep. 2008;57:1079-1080.
poliovirus vaccine followed by oral poliovirus vaccine. Recommendations of 153. Centers for Disease Control and Prevention. Notice to readers: newly licensed
the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm smallpox vaccine to replace old smallpox vaccine. MMWR Morb Mortal Wkly
Rep. 1997;46(RR-3):1-25. Rep. 2008;57:207-208.
2601.e4 PART V  Prevention of Infectious Diseases

154. Centers for Disease Control and Prevention. Syncope after vaccination—United children aged 6-18 years with immunocompromising conditions: recommenda-
States, January 2005-July 2007. MMWR Morb Mortal Wkly Rep. 2008;57:457-460. tions of the Advisory Committee on Immunization Practices (ACIP). MMWR
155. Centers for Disease Control and Prevention. Update: recommendations from the Morb Mortal Wkly Rep. 2013;62:521-524.
Advisory Committee on Immunization Practices (ACIP) regarding administration 177. Centers for Disease Control and Prevention. Use of Japanese encephalitis vaccine
of combination MMRV vaccine. MMWR Morb Mortal Wkly Rep. 2008;57:258-260. in children: recommendations of the advisory committee on immunization
156. Centers for Disease Control and Prevention. Updated recommendations from practices, 2013. MMWR Morb Mortal Wkly Rep. 2013;62:898-900.
the Advisory Committee on Immunization Practices (ACIP) for use of hepatitis 178. Centers for Disease Control and Prevention. Interim guidance for control of serogroup
A vaccine in close contacts of newly arriving international adoptees. MMWR B meningococcal disease outbreaks in organizational settings, 2016. https://www
Morb Mortal Wkly Rep. 2009;58:1006-1007. .cdc.gov/meningococcal/downloads/interim-guidance.pdf.
157. Centers for Disease Control and Prevention. Addition of severe combined 179. Centers for Disease Control and Prevention. Seasonal influenza vaccine safety: a
immunodeficiency as a contraindication for administration of rotavirus vaccine. summary for clinicians, 2016. https://www.cdc.gov/flu/professionals/vaccination/
MMWR Morb Mortal Wkly Rep. 2010;59:687-688. vaccine_safety.htm.
158. Centers for Disease Control and Prevention. Estimates of deaths associated with 180. Chaiken BP, Williams NM, Preblud SR, et al. The effect of a school entry law
seasonal influenza—United States, 1976-2007. MMWR Morb Mortal Wkly Rep. on mumps activity in a school district. JAMA. 1987;257:2455-2458.
2010;59:1057-1062. 181. Chamot E, Toscani L, Egger P, et al. Estimation of the efficacy of three strains
159. Centers for Disease Control and Prevention. FDA licensure of bivalent human of mumps vaccines during an epidemic of mumps in the Geneva canton
papillomavirus vaccine (HPV2, Cervarix) for use in females and updated HPV (Switzerland). Rev Epidemiol Sante Publique. 1998;46:100-107.
vaccination recommendations from the Advisory Committee on Immunization 182. Chang MH, Chen CJ, Lai MS, et al. Universal hepatitis B vaccination in Taiwan
Practices (ACIP). MMWR Morb Mortal Wkly Rep. 2010;59:626-629. and the incidence of hepatocellular carcinoma in children. Taiwan Childhood
160. Centers for Disease Control and Prevention. FDA licensure of quadrivalent Hepatoma Study Group. N Engl J Med. 1997;336:1855-1859.
human papillomavirus vaccine (HPV4, Gardasil) for use in males and guidance 183. Cheek JE, Baron R, Atlas H, et al. Mumps outbreak in a highly vaccinated school
from the Advisory Committee on Immunization Practices (ACIP). MMWR Morb population. Evidence for large-scale vaccination failure. Arch Pediatr Adolesc
Mortal Wkly Rep. 2010;59:630-632. Med. 1995;149:774-778.
161. Centers for Disease Control and Prevention. Invasive pneumococcal disease 184. Chen WH, Cohen MB, Kirkpatrick BD, et al. Single-dose live oral cholera vaccine
in young children before licensure of 13-valent pneumococcal conjugate CVD 103-HgR protects against human experimental infection with Vibrio cholerae
vaccine—United States, 2007. MMWR Morb Mortal Wkly Rep. 2010;59:253-257. O1 El Tor. Clin Infect Dis. 2016;62:1329-1335.
162. Centers for Disease Control and Prevention. Licensure of a 13-valent pneumococcal 185. Chen RT, Glasser JW, Rhodes PH, et al. Vaccine Safety Datalink project: a new
conjugate vaccine (PCV13) and recommendations for use among children - tool for improving vaccine safety monitoring in the United States. The Vaccine
Advisory Committee on Immunization Practices (ACIP), 2010. MMWR Morb Safety Datalink Team. Pediatrics. 1997;99:765-773.
Mortal Wkly Rep. 2010;59:258-261. 186. Chen RT, Moses JM, Markowitz LE, et al. Adverse events following measles-
163. Centers for Disease Control and Prevention. Update: mumps outbreak—New mumps-rubella and measles vaccinations in college students. Vaccine. 1991;9:
York and New Jersey, June 2009-January 2010. MMWR Morb Mortal Wkly Rep. 297-299.
2010;59:125-129. 187. Chen RT, Rastogi SC, Mullen JR, et al. The Vaccine Adverse Event Reporting
164. Centers for Disease Control and Prevention. Update: Recommendations of the System (VAERS). Vaccine. 1994;12:542-550.
Advisory Committee on Immunization Practices (ACIP) regarding use of CSL 188. Cherry JD, Brunell PA, Golden GS, et al. Report of the Task Force on Pertussis
seasonal influenza vaccine (Afluria) in the United States during 2010-11. MMWR and Pertussis Immunization—1988. Pediatrics. 1988;81:933-984.
Morb Mortal Wkly Rep. 2010;59:989-992. 189. Chesson HW, Dunne EF, Hariri S, et al. The estimated lifetime probability
165. Centers for Disease Control and Prevention. Addition of history of intussusception of acquiring human papillomavirus in the United States. Sex Transm Dis.
as a contraindication for rotavirus vaccination. MMWR Morb Mortal Wkly Rep. 2014;41:660-664.
2011;60:1427. 190. Christenson B, Bottiger M. Long-term follow-up study of rubella antibodies in
166. Centers for Disease Control and Prevention. Licensure of a meningococcal naturally immune and vaccinated young adults. Vaccine. 1994;12:41-45.
conjugate vaccine for children aged 2 through 10 years and updated booster dose 191. Christenson B, Bottiger M. Measles antibody: comparison of long-term vaccination
guidance for adolescents and other persons at increased risk for meningococcal titres, early vaccination titres and naturally acquired immunity to and booster
disease—Advisory Committee on Immunization Practices (ACIP), 2011. MMWR effects on the measles virus. Vaccine. 1994;12:129-133.
Morb Mortal Wkly Rep. 2011;60:1018-1019. 192. Chung JR, Flannery B, Thompson MG, et al. Seasonal effectiveness of live
167. Centers for Disease Control and Prevention. Recommendations on the use of attenuated and inactivated influenza vaccine. Pediatrics. 2016;137:e20153279.
quadrivalent human papillomavirus vaccine in males—Advisory Committee 193. Ciofi degli Atti ML, Olin P. Severe adverse events in the Italian and Stockholm
on Immunization Practices (ACIP), 2011. MMWR Morb Mortal Wkly Rep. I pertussis vaccine clinical trials. Dev Biol Stand. 1997;89:77-81.
2011;60:1705-1708. 194. Clements DA, Langdon L, Bland C, et al. Influenza A vaccine decreases the
168. Centers for Disease Control and Prevention. Tetanus surveillance—United States, incidence of otitis media in 6- to 30-month-old children in day care. Arch Pediatr
2001-2008. MMWR Morb Mortal Wkly Rep. 2011;60:365-369. Adolesc Med. 1995;149:1113-1117.
169. Centers for Disease Control and Prevention. Updated recommendations for use 195. Clements DA, Moreira SP, Coplan PM, et al. Postlicensure study of varicella
of meningococcal conjugate vaccines—Advisory Committee on Immunization vaccine effectiveness in a day-care setting. Pediatr Infect Dis J. 1999;18:
Practices (ACIP), 2010. MMWR Morb Mortal Wkly Rep. 2011;60:72-76. 1047-1050.
170. Centers for Disease Control and Prevention. Updated Recommendations for 196. Clifford GM, Smith JS, Plummer M, et al. Human papillomavirus types in invasive
use of tetanus toxoid, reduced diphtheria toxoid and acellular pertussis (Tdap) cervical cancer worldwide: a meta-analysis. Br J Cancer. 2003;88:63-73.
vaccine from the Advisory Committee on Immunization Practices, 2010. MMWR 197. Clover RD, Crawford S, Glezen WP, et al. Comparison of heterotypic protection
Morb Mortal Wkly Rep. 2011;60:13-15. against influenza A/Taiwan/86 (H1N1) by attenuated and inactivated vaccines
171. Centers for Disease Control and Prevention. Updated recommendations for to A/Chile/83-like viruses. J Infect Dis. 1991;163:300-304.
use of tetanus toxoid, reduced diphtheria toxoid and acellular pertussis vaccine 198. Cochi SL, Broome CV, Hightower AW. Immunization of US children with
(Tdap) in pregnant women and persons who have or anticipate having close Hemophilus influenzae type b polysaccharide vaccine. A cost-effectiveness model
contact with an infant aged <12 months— Advisory Committee on Immunization of strategy assessment. JAMA. 1985;253:521-529.
Practices (ACIP), 2011. MMWR Morb Mortal Wkly Rep. 2011;60:1424-1426. 199. Cochi SL, Preblud SR, Orenstein WA. Prespectives on the relative resurgence of
172. Centers for Disease Control and Prevention. Use of hepatitis B vaccination mumps in the United States. Am J Dis Child. 1988;142:499-507.
for adults with diabetes mellitus: recommendations of the Advisory Com- 200. Cody CL, Baraff LJ, Cherry JD, et al. Nature and rates of adverse reactions
mittee on Immunization Practices (ACIP). MMWR Morb Mortal Wkly Rep. associated with DTP and DT immunizations in infants and children. Pediatrics.
2011;60:1709-1711. 1981;68:650-660.
173. Centers for Disease Control and Prevention. Mumps outbreak on a university 201. Cohen R. Impact of 13-valent pneumococcal conjugate vaccine on pneumococcal
campus—California, 2011. MMWR Morb Mortal Wkly Rep. 2012;61:986-989. nasopharyngeal carriage in children with acute otitis media. Pediatr Infect Dis
174. Centers for Disease Control and Prevention. Progress in introduction of pneu- J. 2012;31:297.
mococcal conjugate vaccine–worldwide, 2000-2012. MMWR Morb Mortal Wkly 202. Cohen C, White JM, Savage EJ, et al. Vaccine effectiveness estimates, 2004-2005
Rep. 2013;62:308-311. mumps outbreak, England. Emerg Infect Dis. 2007;13:12-17.
175. Centers for Disease Control and Prevention. Updated recommendations for 203. Cohn A Proceedings of the Advisory Committee on Immunization Practices (ACIP)
use of tetanus toxoid, reduced diphtheria toxoid, and acellular pertussis vaccine meeting. Paper presented at: Advisory Committee on Immunization Practices
(Tdap) in pregnant women—Advisory Committee on Immunization Practices (ACIP) meeting; 2010, Atlanta, GA.
(ACIP), 2012. MMWR Morb Mortal Wkly Rep. 2013;62:131-135. 204. Cohn AC, MacNeil JR, Clark TA, et al. Prevention and control of meningococcal
176. Centers for Disease Control and Prevention. Use of 13-valent pneumococcal disease: recommendations of the Advisory Committee on Immunization Practices
conjugate vaccine and 23-valent pneumococcal polysaccharide vaccine among (ACIP). MMWR Recomm Rep. 2013;62(RR-2):1-28.
CHAPTER 245  Active Immunizing Agents 2601.e5

205. Cohn AC, MacNeil JR, Harrison LH, et al. Changes in Neisseria meningitidis 234. Dennehy PH, Bertrand HR, Silas PE, et al. Coadministration of RIX4414
disease epidemiology in the United States, 1998-2007: implications for prevention oral human rotavirus vaccine does not impact the immune response to
of meningococcal disease. Clin Infect Dis. 2010;50:184-191. antigens contained in routine infant vaccines in the United States. Pediatrics.
206. Cohn ML, Robinson ED, Faerber M, et al. Measles vaccine failures: lack of sustained 2008;122:e1062-e1066.
measles-specific immunoglobulin G responses in revaccinated adolescents and 235. Dennehy PH, Brady RC, Halperin SA, et al. Comparative evaluation of safety
young adults. Pediatr Infect Dis J. 1994;13:34-38. and immunogenicity of two dosages of an oral live attenuated human rotavirus
207. Colditz GA, Berkey CS, Mosteller F, et al. The efficacy of bacillus Calmette- vaccine. Pediatr Infect Dis J. 2005;24:481-488.
Guerin vaccination of newborns and infants in the prevention of tuberculosis: 236. Dennehy PH, Goveia MG, Dallas MJ, et al. The integrated phase III safety profile
meta-analyses of the published literature. Pediatrics. 1995;96:29-35. of the pentavalent human-bovine (WC3) reassortant rotavirus vaccine. Int J
208. Collins S, Mazloomzadeh S, Winter H, et al. High incidence of cervical human Infect Dis. 2007;11(suppl 2):S36-S42.
papillomavirus infection in women during their first sexual relationship. BJOG. 237. Dennehy PH, Vesikari T, Matson DO, et al. Efficacy of the pentavalent rotavirus
2002;109:96-98. vaccine, RotaTeq® (RV5), between doses of a 3-dose series and with less than
209. Committee on Infectious Diseases, American Academy of Pediatrics. HPV vaccine 3 doses (incomplete regimen). Hum Vaccin. 2011;7:563-568.
recommendations. Pediatrics. 2012;129:602-605. 238. Denoel PA, Goldblatt D, de Vleeschauwer I, et al. Quality of the Haemophilus
210. Committee on Infectious Diseases, American Academy of Pediatrics. Recommended influenzae type b (Hib) antibody response induced by diphtheria-tetanus-acellular
childhood and adolescent immunization schedule—United States, 2016. Pediatrics. pertussis/Hib combination vaccines. Clin Vaccine Immunol. 2007;14:1362-1369.
2016;137:e20154531. 239. D’Hondt E. Possible approaches to develop vaccines against hepatitis A. Vaccine.
211. Committee on Infectious Diseases American Academy of Pediatrics. Recom- 1992;10:S48-S52.
mendations for serogroup B meningococcal vaccine for persons 10 years and 240. Dominguez A, Salleras L, Carmona G, et al. Effectiveness of a mass hepatitis A
older. Pediatrics. 2016;138(3):pii: e20161890. vaccination program in preadolescents. Vaccine. 2003;21:698-701.
212. Committee on Issues and Priorities for New Vaccine Development. New Vaccine 241. Dominguez A, Torner N, Castilla J, et al. Mumps vaccine effectiveness in highly
Develoment: Establishing Priorities. Vol. I. Diseases of Importance in the United immunized populations. Vaccine. 2010;28:3567-3570.
States. Washington, DC: National Academy Press; 1985. 242. Donahue JG, Choo PW, Manson JE, et al. The incidence of herpes zoster. Arch
213. Committee on Issues and Priorities for New Vaccine Development. New Vaccine Intern Med. 1995;155:1605-1609.
Develoment: Establishing Priorities. Vol. II. Diseases of Importance in Developing 243. Donegan K, King B, Bryan P. Safety of pertussis vaccination in pregnant women
Countries. Washington, DC: National Academy Press; 1986. in UK: observational study. BMJ. 2014;349:g4219.
214. Committee to Study Priorities for Vaccine Development, Institute of Medicine. 244. Donovan B, Franklin N, Guy R, et al. Quadrivalent human papillomavirus vac-
Vaccines for the 21st Century: A Tool for Decision Making. Washington, DC: cination and trends in genital warts in Australia: analysis of national sentinel
National Academies Press; 2000. surveillance data. Lancet Infect Dis. 2011;11:39-44.
215. Cortes JE, Curns AT, Tate JE, et al. Rotavirus vaccine and health care utilization 245. Donovan B, Grulich AE. The quadrivalent HPV vaccine is effective prophylaxis
for diarrhea in U.S. children. N Engl J Med. 2011;365:1108-1117. against HPV-related external genital lesions in young men. Evid Based Med.
216. Cortese MM, Jordan HT, Curns AT, et al. Mumps vaccine performance among 2011;16:157-158.
university students during a mumps outbreak. Clin Infect Dis. 2008;46:1172-1180. 246. Douvoyiannis M, Belamarich PF, Goldman DL. Tetanus after vaccine refusal
217. Cortese MM, Parashar UD. Prevention of rotavirus gastroenteritis among infants and an opportunity for the pediatric infectious diseases specialist. Clin Pediatr
and children: recommendations of the Advisory Committee on Immunization (Phila). 2015;54:513-516.
Practices (ACIP). MMWR Recomm Rep. 2009;58(RR-2):1-25. 247. Dubischar-Kastner K, Eder S, Buerger V, et al. Long-term immunity and immune
218. Coulehan JL, Michaels RH, Hallowell C, et al. Epidemiology of Haemophilus influ- response to a booster dose following vaccination with the inactivated Japanese
enzae type B disease among Navajo Indians. Public Health Rep. 1984;99:404-409. encephalitis vaccine IXIARO, IC51. Vaccine. 2010;28:5197-5202.
219. Crossley K, Irvine P, Warren JB, et al. Tetanus and diphtheria immunity in urban 248. Dubischar-Kastner K, Kaltenboeck A, Klingler A, et al. Safety analysis of a Vero-cell
Minnesota adults. JAMA. 1979;242:2298-2300. culture derived Japanese encephalitis vaccine, IXIARO (IC51), in 6 months of
220. Cutts FT, Zaman SM, Enwere G, et al. Efficacy of nine-valent pneumococcal follow-up. Vaccine. 2010;28:6463-6469.
conjugate vaccine against pneumonia and invasive pneumococcal disease in 249. Dunne EF, Nielson CM, Stone KM, et al. Prevalence of HPV infection among
The Gambia: randomised, double-blind, placebo-controlled trial. Lancet. men: a systematic review of the literature. J Infect Dis. 2006;194:1044-1057.
2005;365:1139-1146. 250. Duvriendt J, Staroukine MM, Costy F, et al. Fatal encephalitis apparently due
221. Dagan R, Amir J, Mijalovsky A, et al. Immunization against hepatitis A in the to rabies: Occurrence after treatment with human diploid cell vaccine but not
first year of life: priming despite the presence of maternal antibody. Pediatr rabies immune globulin. JAMA. 1982;248:2304-2306.
Infect Dis J. 2000;19:1045-1052. 251. Dworkin MS, Jennings CE, Roth-Thomas J, et al. An outbreak of varicella among
222. Dagan R, Givon-Lavi N, Fraser D, et al. Serum serotype-specific pneumococcal children attending preschool and elementary school in Illinois. Clin Infect Dis.
anticapsular immunoglobulin g concentrations after immunization with a 9-valent 2002;35:102-104.
conjugate pneumococcal vaccine correlate with nasopharyngeal acquisition of 252. Edwards KM, Decker MD. Acellular pertussis vaccines for infants. N Engl J Med.
pneumococcus. J Infect Dis. 2005;192:367-376. 1996;334:391-392.
223. Dagan R, Melamed R, Muallem M, et al. Reduction of nasopharyngeal car- 253. Edwards KM, Meade BD, Decker MD, et al. Comparison of 13 acellular pertussis
riage of pneumococci during the second year of life by a heptavalent conjugate vaccines: overview and serologic response. Pediatrics. 1995;96:548-557.
pneumococcal vaccine. J Infect Dis. 1996;174:1271-1278. 254. Ehrenkranz NJ, Ventura AK, Medler EM, et al. Clinical evaluation of a new
224. Dagan R, Muallem M, Melamed R, et al. Reduction of pneumococcal nasopharyn- measles-mumps-rubella combined live virus vaccine in the Dominican Republic.
geal carriage in early infancy after immunization with tetravalent pneumococcal Bull World Health Organ. 1975;52:81-85.
vaccines conjugated to either tetanus toxoid or diphtheria toxoid. Pediatr Infect 255. Eisenberg KW, Szilagyi PG, Fairbrother G, et al. Vaccine effectiveness against
Dis J. 1997;16:1060-1064. laboratory-confirmed influenza in children 6 to 59 months of age during the
225. D’Amelio R, Biselli R, Fascia G, et al. Measles-mumps-rubella vaccine in the 2003-2004 and 2004-2005 influenza seasons. Pediatrics. 2008;122:911-919.
Italian armed forces. JAMA. 2000;284:2059. 256. Ellis RW, Rappuoli R, Ahmed S. New technologies for making vaccines. In:
226. Daubeney P, Taylor CJ, McGaw J, et al. Immunogenicity and tolerability of a Plotkin S, Orenstein W, Offit P, eds. Vaccines. 6th ed. Philadelphia, PA: Saunders
trivalent influenza subunit vaccine (Influvac) in high-risk children aged 6 months Elsevier; 2013:1182-1199.
to 4 years. Br J Clin Pract. 1997;51:87-90. 257. Englund JA, Walter EB, Fairchok MP, et al. A comparison of 2 influenza vaccine
227. Davidkin I, Jokinen S, Broman M, et al. Persistence of measles, mumps, and schedules in 6- to 23-month-old children. Pediatrics. 2005;115:1039-1047.
rubella antibodies in an MMR-vaccinated cohort: a 20-year follow-up. J Infect 258. Englund JA, Walter EB, Gbadebo A, et al. Immunization with trivalent inactivated
Dis. 2008;197:950-956. influenza vaccine in partially immunized toddlers. Pediatrics. 2006;118:e579-e585.
228. Davidson M, Schraer CD, Parkinson AJ, et al. Invasive pneumococcal disease in 259. Eskola J, Anttila M. Pneumococcal conjugate vaccines. Pediatr Infect Dis J.
an Alaska native population, 1980 through 1986. JAMA. 1989;261:715-718. 1999;18:543-551.
229. Dayan GH, Quinlisk MP, Parker AA, et al. Recent resurgence of mumps in the 260. Eskola J, Kilpi T, Palmli A, et al. Efficacy of a pneumococcal conjugate vaccine
United States. N Engl J Med. 2008;358:1580-1589. against otitis media. N Engl J Med. 2001;344:430-439.
230. Decker MD, Edwards KM. Haemophilus influenzae type b vaccines: history, choice 261. Euler GL, Wooten KG, Baughman AL, et al. Hepatitis B surface antigen prevalence
and comparisons. Pediatr Infect Dis J. 1998;17(9 suppl):S113-S116. among pregnant women in urban areas: implications for testing, reporting, and
231. Decker MD, Edwards KM. Report of the nationwide multicenter acellular pertussis preventing perinatal transmission. Pediatrics. 2003;111:1192-1197.
trial. Pediatrics. 1995;96:547-603. 262. Evans G. National Childhood Vaccine Injury Act: revision of the vaccine injury
232. Decker MD, Edwards KM, Bradley R, et al. Comparative trial in infants of four table. Pediatrics. 1996;98:1179-1181.
conjugate Haemophilus influenzae type b vaccines. J Pediatr. 1992;120:184-189. 263. Feikin DR, Kagucia EW, Loo JD, et al. Serotype-specific changes in invasive
233. Deeks SL, Lim GH, Simpson MA, et al. An assessment of mumps vaccine pneumococcal disease after pneumococcal conjugate vaccine introduction: a
effectiveness by dose during an outbreak in Canada. CMAJ. 2011;183:1014-1020. pooled analysis of multiple surveillance sites. PLoS Med. 2013;10:e1001517.
2601.e6 PART V  Prevention of Infectious Diseases

264. Feiterna-Sperling C, Bronnimann R, Tischer A, et al. Open randomized trial 292. Gershon A, Takahashi M, Seward J. Live attenuated varicella vaccine. In: Plotkin
comparing the immunogenicity and safety of a new measles-mumps-rubella S, Orenstein W, Offit P, eds. Vaccines. 5th ed. Philadelphia: Sanders Elsevier;
vaccine and a licensed vaccine in 12- to 24-month-old children. Pediatr Infect 2008:915-958.
Dis J. 2005;24:1083-1088. 293. Ghaffar F, Barton T, Lozano J, et al. Effect of the 7-valent pneumococcal conjugate
265. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality vaccine on nasopharyngeal colonization by Streptococcus pneumoniae in the first
worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J 2 years of life. Clin Infect Dis. 2004;39:930-938.
Cancer. 2015;136:E359-E386. 294. Giebink GS. Preventing pneumococcal disease in children: recommendations
266. Fiebelkorn A, Barskey A, Hickman C, Centers for Disease Control and Prevention, for using pneumococcal vaccine. Pediatr Infect Dis. 1985;4:343-348.
et al. Mumps. In: Manual for the Surveillance of Vaccine-Preventable Diseases. 5th 295. Giebink GS, Le CT, Schiffman G. Decline of serum antibody in splenectomized
ed. Atlanta, GA: US Department of Health and Human Services, Centers for children after vaccination with pneumococcal capsular polysaccharides. J Pediatr.
Disease Control and Prevention; 2012. 1984;105:576-582.
267. Fiebelkorn AP, Redd SB, Gastanaduy PA, et al. A comparison of postelimination 296. Gill CJ, Baxter R, Anemona A, et al. Persistence of immune responses after a
measles epidemiology in the United States, 2009-2014 versus 2001-2008. J Pediatric single dose of Novartis meningococcal serogroup A, C, W-135 and Y CRM-197
Infect Dis Soc. 2017;6(1):40-48. conjugate vaccine (Menveo® or Menactra®) among healthy adolescents. Hum
268. Fine PEM, Clarkson JA. Reflections on the efficacy of pertussis vaccine. Rev Vaccin. 2010;6:881-887.
Infect Dis. 1987;9:866-883. 297. Gilliland SM, Forrest L, Carre H, et al. Investigation of porcine circovirus
269. Fiore AE, Shapiro CN, Sabin K, et al. Hepatitis A vaccination of infants: effect contamination in human vaccines. Biologicals. 2012;40:270-277.
of maternal antibody status on antibody persistence and response to a booster 298. Glass RI, Kilgore PE, Holman RC, et al. The epidemiology of rotavirus diarrhea
dose. Pediatr Infect Dis J. 2003;22:354-359. in the United States: surveillance and estimates of disease burden. J Infect Dis.
270. Fiore AE, Wasley A, Bell BP. Prevention of hepatitis A through active or passive 1996;174(suppl 1):S5-S11.
immunization: recommendations of the Advisory Committee on Immunization 299. Glezen WP. Serious morbidity and mortality associated with influenza epidemics.
Practices (ACIP). MMWR Recomm Rep. 2006;55(RR-7):1-23. Epidemiol Rev. 1982;4:25-44.
271. Fischer M, Lindsey N, Staples JE, et al. Japanese encephalitis vaccines: recom- 300. Glezen WP, Couch RB. Interpandemic influenza in the Houston area, 1974-76.
mendations of the Advisory Committee on Immunization Practices (ACIP). N Engl J Med. 1978;298:587-592.
MMWR Recomm Rep. 2010;59(RR-1):1-27. 301. Glezen WP, Greenberg SB, Atmar RL, et al. Impact of respiratory virus infections
272. Fishbein DB, Sawyer LA, Reid-Sanden FL, et al. Administration of human diploid- on persons with chronic underlying conditions. JAMA. 2000;283:499-505.
cell rabies vaccine in the gluteal area [letter]. N Engl J Med. 1988;318:124-125. 302. Glezen W, Six H, Frank A, et al. Impact of epidemics upon communities and
273. Fishbein DB, Yenne KM, Dreesen DW, et al. Risk factors for systemic hypersensitiv- families. In: Kendal A, Patriaca P, eds. Options for the Control of Influenza. New
ity reactions after booster vaccinations with human diploid cell rabies vaccine: York: Alan R Liss; 1986:63-73.
a nationwide prospective study. Vaccine. 1993;11:1390-1394. 303. Gold R, Lepow ML, Goldschneider I, et al. Kinetics of antibody production
274. Fletcher HA, Schrager L. TB vaccine development and the End TB Strategy: to group A and group C meningococcal polysaccharide vaccines administered
importance and current status. Trans R Soc Trop Med Hyg. 2016;110:212-218. during the first six years of life: prospects for routine immunization of infants
275. Fogel A, Moshkowitz A, Rannon L, et al. Comparative trials of RA 27-3 and and children. J Infect Dis. 1979;140:690-697.
Cendehill rubella vaccines in adult and adolescent females. Am J Epidemiol. 304. Goldschneider I, Gotschlich EC, Artenstein MS. Human immunity to the
1971;93:392-398. meningococcus. I. The role of humoral antibodies. J Exp Med. 1969;129:1307-1326.
276. Folaranmi T, Rubin L, Martin SW, et al. Use of serogroup B meningococcal vaccines 305. Gonzalez M, Pirez MC, Ward E, et al. Safety and immunogenicity of a paediatric
in persons aged ≥10 years at increased risk for serogroup B meningococcal disease: presentation of an influenza vaccine. Arch Dis Child. 2000;83:488-491.
recommendations of the Advisory Committee on Immunization Practices, 2015. 306. Gothefors L, Bergstrom E, Backman M. Immunogenicity and reactogenicity of
MMWR Morb Mortal Wkly Rep. 2015;64:608-612. a new measles, mumps and rubella vaccine when administered as a second dose
277. Food and Drug Administration. December 10, 2014 Approval Letter—Gardasil at 12 y of age. Scand J Infect Dis. 2001;33:545-549.
9. Silver Spring, MD: US Department of Health and Human Services; 2014. 307. Gotschlich EC, Goldschneider I, Artenstein MS. Human immunity to the
278. Food and Drug Administration. October 7, 2016 Approval Letter for Human meningococcus. IV. Immunogenicity of group A and group C meningococcal
Papillomavirus 9-Valent Vaccine, Recombinant to Include a 2-Dose Regimen for polysaccharides in human volunteers. J Exp Med. 1969;129:1367-1384.
Individuals 9 Through 14 Years of Age. Silver Spring, MD: US Department of 308. Grassly NC. Immunogenicity and effectiveness of routine immunization with 1
Health and Human Services; 2016. or 2 doses of inactivated poliovirus vaccine: systematic review and meta-analysis.
279. Fowlkes A, Steffens A, Temte J, et al. Incidence of medically attended influenza J Infect Dis. 2014;210(suppl 1):S439-S446.
during pandemic and post-pandemic seasons through the Influenza Incidence 309. Greco D, Salmaso S, Mastrantonio P, et al. A controlled trial of two acellular
Surveillance Project, 2009-13. Lancet Respir Med. 2015;3:709-718. vaccines and one whole-cell vaccine against pertussis. Progetto Pertosse Working
280. Freed GL, Katz SL, Clark SJ. Safety of vaccinations. Miss America, the media, Group. N Engl J Med. 1996;334:341-348.
and public health. JAMA. 1996;276:1869-1872. 310. Greenberg DP. Pediatric experience with recombinant hepatitis B vaccines and
281. Freestone DS, Reynolds GM, McKinnon JA, et al. Vaccination of schoolgirls relevant safety and immunogenicity studies. Pediatr Infect Dis J. 1993;12:438-445.
against rubella. Assessment of serological status and a comparative trial of Wistar 311. Greenberg MA, Birx DL. Safe administration of mumps-measles-rubella vaccine
RA 27/3 and Cendehill strain live attenuated rubella vaccines in 13-year-old in egg-allergic children. J Pediatr. 1988;113:504-506.
schoolgirls in Dudley. Br J Prev Soc Med. 1975;29:258-261. 312. Greenberg DP, Lieberman JM, Marcy SM, et al. Enhanced antibody responses
282. Fritzell B, Plotkin S. Efficacy and safety of a Haemophilus influenzae type b in infants given different sequences of heterogeneous Haemophilus influenzae
capsular polysaccharide-tetanus protein conjugate vaccine. J Pediatr. 1992;121: type b conjugate vaccines. J Pediatr. 1995;126:206-211.
355-362. 313. Greene SK, Kulldorff M, Lewis EM, et al. Near real-time surveillance for influenza
283. Furukawa T, Miyata T, Kondo K, et al. Rubella vaccination during an epidemic. vaccine safety: proof-of-concept in the Vaccine Safety Datalink Project. Am J
JAMA. 1970;213:987-990. Epidemiol. 2010;171:177-188.
284. Gaglani M, Pruszynski J, Murthy K, et al. Influenza vaccine effectiveness against 314. Grillner L. Neutralizing antibodies after rubella vaccination of newly delivered
2009 pandemic influenza A(H1N1) virus differed by vaccine type during 2013-2014 women: a comparison between three vaccines. Scand J Infect Dis. 1975;7:169-172.
in the United States. J Infect Dis. 2016;213:1546-1556. 315. Grohskopf LA, Sokolow LZ, Broder KR, et al. Prevention and control of seasonal
285. Galazka AM, Robertson SE. Diphtheria: changing patterns in the developing influenza with vaccines. MMWR Recomm Rep. 2016;65:1-54.
world and the industrialized world. Eur J Epidemiol. 1995;11:107-117. 316. Groothuis JR, Lehr MV, Levin MJ. Safety and immunogenicity of a purified
286. Galil K, Fair E, Mountcastle N, et al. Younger age at vaccination may increase haemagglutinin antigen in very young high-risk children. Vaccine. 1994;12:139-141.
risk of varicella vaccine failure. J Infect Dis. 2002;186:102-105. 317. Gross PA, Russo C, Dran S, et al. Time to earliest peak serum antibody response
287. Gastanaduy PA, Redd SB, Fiebelkorn AP, et al. Measles—United States, January to influenza vaccine in the elderly. Clin Diagn Lab Immunol. 1997;4:491-492.
1-May 23, 2014. MMWR Morb Mortal Wkly Rep. 2014;63:496-499. 318. Group GA. Expanded programme on immunization. Global Advisory Group—Part
288. Gatchalian S, Cordero-Yap L, Lu-Fong M, et al. A randomized comparative trial II. Achieving the major disease control goals. Wkly Epidemiol Rec. 1994;69:29-31,
in order to assess the reactogenicity and immunogenicity of a new measles 34-35.
mumps rubella (MMR) vaccine when given as a first dose at 12-24 months of 319. Guerra FA, Blatter MM, Greenberg DP, et al. Safety and immunogenicity of a
age. Southeast Asian J Trop Med Public Health. 1999;30:511-517. pentavalent vaccine compared with separate administration of licensed equivalent
289. Gergen PJ, McQuillan GM, Kiely M, et al. A population-based serologic survey vaccines in US infants and toddlers and persistence of antibodies before a preschool
of immunity to tetanus in the United States. N Engl J Med. 1995;332:761-766. booster dose: a randomized, clinical trial. Pediatrics. 2009;123:301-312.
290. Gershon AA, LaRussa PS. Varicella vaccine. Pediatr Infect Dis J. 1998;17: 320. Guess HA, Broughton DD, Melton I, et al. Population-based studies of varicella
248-249. complications. Pediatrics. 1986;78(suppl):723-727.
291. Gershon AA, Steinberg SP, LaRussa P, et al. Immunization of healthy adults with 321. Gupta RK, Griffin P Jr, Xu J, et al. Diphtheria antitoxin levels in US blood and
ive attenuated varicella vaccine. J Infect Dis. 1988;158:132-137. plasma donors. J Infect Dis. 1996;173:1493-1497.
CHAPTER 245  Active Immunizing Agents 2601.e7

322. Guris D, Jumaan AO, Mascola L, et al. Changing varicella epidemiology in 352. Holmes SJ, Fritzell B, Guito KP, et al. Immunogenicity of Hemophilus influenzae
active surveillance sites—United States, 1995-2005. J Infect Dis. 2008;197(suppl type b polysaccharide-tetanus toxoid conjugate vaccine in infants. Am J Dis
2):S71-S75. Child. 1993;147:832-836.
323. Gustafsson L, Hallander HO, Olin P, et al. A controlled trial of a two-component 353. Horstmann DM, Schluederberg A, Emmons JE, et al. Persistence of vaccine-induced
acellular, a five-component acellular, and a whole-cell pertussis vaccine. N Engl immune responses to rubella: comparison with natural infection. Rev Infect Dis.
J Med. 1996;334:349-355. 1985;7(suppl 1):S80-S85.
324. Guzman CA, Borsutzky S, Griot-Wenk M, et al. Vaccines against typhoid fever. 354. Howson CP, Katz M, Johnston RB Jr, et al. Chronic arthritis after rubella vac-
Vaccine. 2006;24:3804-3811. cination. Clin Infect Dis. 1992;15:307-312.
325. Haber P, Patel M, Izurieta HS, et al. Postlicensure monitoring of intussusception 355. Hsieh YC, Wu FT, Hsiung CA, et al. Comparison of virus shedding after
after RotaTeq vaccination in the United States, February 1, 2006, to September lived attenuated and pentavalent reassortant rotavirus vaccine. Vaccine.
25, 2007. Pediatrics. 2008;121:1206-1212. 2014;32:1199-1204.
326. Hadler SC, McFarland L. Hepatitis in day care centers: epidemiology and 356. Hsu HY, Chang MH, Liaw SH, et al. Changes of hepatitis B surface antigen
prevention. Rev Infect Dis. 1986;8:548-557. variants in carrier children before and after universal vaccination in Taiwan.
327. Halperin SA, Gupta A, Jeanfreau R, et al. Comparison of the safety and immu- Hepatology. 1999;30:1312-1317.
nogenicity of an investigational and a licensed quadrivalent meningococcal 357. Ildirim I, Hacimustafaoglu M, Ediz B. Correlation of tuberculin induration
conjugate vaccine in children 2-10 years of age. Vaccine. 2010;28:7865-7872. with the number of Bacillus Calmette-Guérin vaccines. Pediatr Infect Dis J.
328. Halsey NA, Hyman SL. Measles-mumps-rubella vaccine and autistic spectrum 1995;14:1060-1063.
disorder: report from the New Challenges in Childhood Immunizations Conference 358. Innis BL, Snitbhan R, Kunasol P, et al. Protection against hepatitis A by an
convened in Oak Brook, Illinois, June 12-13, 2000. Pediatrics. 2001;107:E84. inactivated vaccine. JAMA. 1994;271:1328-1334.
329. Hambidge SJ, Glanz JM, France EK, et al. Safety of trivalent inactivated influenza 359. Institute of Medicine. Diphtheria and tetanus toxoids. In: Stratton KR, Howe CJ,
vaccine in children 6 to 23 months old. JAMA. 2006;296:1990-1997. Johnston RB Jr, eds. Adverse Events Associated with Childhood Vaccines: Evidence
330. Hammerschlag M, Gershon A, Steinberg S, et al. Herpes zoster in an adult recipient Bearing on Causality. Washington, DC: National Academy Press; 1994.
of live attenuated varicella vaccine. J Infect Dis. 1989;160:535-537. 360. Institute of Medicine. Immunization Safety Review: Measles-Mumps-Rubella
331. Hampton LM, Farley MM, Schaffner W, et al. Prevention of antibiotic- Vaccine and Autism. Washington, DC: National Academy Press; 2001.
nonsusceptible Streptococcus pneumoniae with conjugate vaccines. J Infect Dis. 361. Institute of Medicine. Immunization Safety Review: Vaccines and Autism.
2012;205:401-411. Washington, DC: National Academy Press; 2004.
332. Hanna-Wakim R, Yasukawa LL, Sung P, et al. Immune responses to mumps 362. Institute of Medicine. Measles and mumps vaccine. In: Stratton KR, Howe CJ,
vaccine in adults who were vaccinated in childhood. J Infect Dis. 2008;197: Johnston RB Jr, eds. Adverse Events Associated with Childhood Vaccines. Washington
1669-1675. DC: National Academy Press; 1994:118-186.
333. Hansen J, Black S, Shinefield H, et al. Effectiveness of heptavalent pneumococcal 363. Ipp MM, Gold R, Goldbach M, et al. Adverse reactions to diphtheria, tetanus,
conjugate vaccine in children younger than 5 years of age for prevention of pertussis-polio vaccination at 18 months of age: effect of injection site and
pneumonia: updated analysis using World Health Organization standardized needle length. Pediatrics. 1989;83:679-682.
interpretation of chest radiographs. Pediatr Infect Dis J. 2006;25:779-781. 364. Isaacman DJ, McIntosh ED, Reinert RR. Burden of invasive pneumococcal disease
334. Hardy IR, Dittmann S, Sutter RW. Current situation and control strategies for and serotype distribution among Streptococcus pneumoniae isolates in young
resurgence of diphtheria in newly independent states of the former Soviet Union. children in Europe: impact of the 7-valent pneumococcal conjugate vaccine and
Lancet. 1996;347:1739-1744. considerations for future conjugate vaccines. Int J Infect Dis. 2010;14:e197-e209.
335. Harling R, White JM, Ramsay ME, et al. The effectiveness of the mumps component 365. Izurieta HS, Strebel PM, Blake PA. Postlicensure effectiveness of varicella vaccine
of the MMR vaccine: a case control study. Vaccine. 2005;23:4070-4074. during an outbreak in a childcare center. JAMA. 1997;278:1495-1499.
336. Harper DM, Franco EL, Wheeler C, et al. Efficacy of a bivalent L1 virus-like particle 366. Jackson BR, Iqbal S, Mahon B, et al. Updated recommendations for the use
vaccine in prevention of infection with human papillomavirus types 16 and 18 of typhoid vaccine—Advisory Committee on Immunization Practices, United
in young women: a randomised controlled trial. Lancet. 2004;364:1757-1765. States, 2015. MMWR Morb Mortal Wkly Rep. 2015;64:305-308.
337. Hart PDA, Sutherland I, Thomas J. The immunity conferred by effective BCG 367. Jacobs RL, Lowe RS, Lanier BQ. Adverse reactions to tetanus toxoid. JAMA.
and vole bacillus vaccines, in relation to individual variations in induced tuber- 1982;247:40-42.
culin sensitivity and to technical variations in the vaccines. Tubercle. 1967;48: 368. Jafri RZ, Ali A, Messonnier NE, et al. Global epidemiology of invasive meningococ-
201-210. cal disease. Popul Health Metr. 2013;11:17.
338. Heath PT, McVernon J. The UK Hib vaccine experience. Arch Dis Child. 369. James JM, Burks AW, Roberson PK, et al. Safe administration of the measles
2002;86:396-399. vaccine to children allergic to eggs. N Engl J Med. 1995;332:1262-1266.
339. Heikkinen T, Ruuskanen O, Waris M, et al. Influenza vaccination in the prevention 370. Jefferson T, Rivetti A, Harnden A, et al. Vaccines for preventing influenza in
of acute otitis media in children. Am J Dis Child. 1991;145:445-448. healthy children. Cochrane Database Syst Rev. 2008;(2):CD004879.
340. Herman JJ, Radin R, Schneiderman R. Allergic reactions to measles (rubeola) 371. Jhung MA, Swerdlow D, Olsen SJ, et al. Epidemiology of 2009 pandemic influenza
vaccine in patients hypersensitive to egg protein. J Pediatr. 1983;102: A (H1N1) in the United States. Clin Infect Dis. 2011;52(suppl 1):S13-S26.
196-199. 372. Johnson CE, Kumar ML, Whitwell JK, et al. Antibody persistence after primary
341. Hersh BS, Fine PE, Kent WK, et al. Mumps outbreak in a highly vaccinated measles-mumps-rubella vaccine and response to a second dose given at four to
population. J Pediatr. 1991;119:187-193. six vs. eleven to thirteen years. Pediatr Infect Dis J. 1996;15:687-692.
342. Hessel L, Debois H, Fletcher M, et al. Experience with Salmonella typhi Vi capsular 373. Johnson KJ, Gallagher NM, Mintz ED, et al. From the CDC: new country-
polysaccharide vaccine. Eur J Clin Microbiol Infect Dis. 1999;18:609-620. specific recommendations for pre-travel typhoid vaccination. J Travel Med.
343. Hetherington S, Lepow ML. Epidemiology and immunology of Hemophilus 2011;18:430-433.
influenzae type B infections in childhood: implications for chemoprophylaxis 374. Johnson KJ. Crossing borders: one world, global health: CDC updates recom-
and immunization. Adv Pediatr Infect Dis. 1987;2:1-18. mendations for typhoid vaccination. Clin Infect Dis. 2012;54:v-vi.
344. Hill HA, Elam-Evans LD, Yankey D, et al. Vaccination coverage among children 375. Johnson MG, Bradley KK, Mendus S, et al. Vaccine-preventable disease
aged 19-35 months—United States, 2015. MMWR Morb Mortal Wkly Rep. among homeschooled children: two cases of tetanus in Oklahoma. Pediatrics.
2016;65:1065-1071. 2013;132:e1686-e1689.
345. Hill HA, Elam-Evans LD, Yankey D, et al. Vaccination coverage among children 376. Johnson CE, Stancin T, Fattlar D, et al. A long-term prospective study of varicella
aged 19-35 months—United States, 2015. MMWR Morb Mortal Wkly Rep. vaccine in healthy children. Pediatrics. 1997;100:761-766.
2016;65:1065-1071. 377. Jokinen S, Osterlund P, Julkunen I, et al. Cellular immunity to mumps virus
346. Deleted in review. in young adults 21 years after measles-mumps-rubella vaccination. J Infect Dis.
347. Hilleman MR, Buynak EB, Weibel RE, et al. Live, attenuated mumps-virus vaccine. 2007;196:861-867.
N Engl J Med. 1968;278:227-232. 378. Joura EA, Giuliano AR, Iversen OE, et al. A 9-valent HPV vaccine against infection
348. Hilleman MR, Weibel RE, Buynak EB, et al. Live attenuated mumps-virus and intraepithelial neoplasia in women. N Engl J Med. 2015;372:711-723.
vaccine. IV. Protective efficacy as measured in a field evaluation. N Engl J Med. 379. Kahn JA, Widdice LE, Ding L, et al. Substantial decline in vaccine-type human
1967;276:252-258. papillomavirus (HPV) among vaccinated young women during the first 8 years
349. Hinman AR, Koplan JP. Pertussis and pertussis vaccine: reanalysis of benefits, after HPV vaccine introduction in a community. Clin Infect Dis. 2016;63:1281-1287.
risks and costs. JAMA. 1984;251:3109-3113. 380. Kanra G, Yalcin SS, Kara A, et al. Hepatitis A booster vaccine in children after
350. Ho GY, Burk RD, Klein S, et al. Persistent genital human papillomavirus infection as infant immunization. Pediatr Infect Dis J. 2002;21:727-730.
a risk factor for persistent cervical dysplasia. J Natl Cancer Inst. 1995;87:1365-1371. 381. Kaplan SL. Streptococcus pneumoniae: impact of antibiotic resistance in pediatrics.
351. Hoberman A, Greenberg DP, Paradise JL, et al. Effectiveness of inactivated influenza Curr Probl Pediatr. 1997;27:187-195.
vaccine in preventing acute otitis media in young children: a randomized controlled 382. Katz SL, Hinman AR. Summary and conclusions: measles elimination meeting,
trial. JAMA. 2003;290:1608-1616. 16-17 March 2000. J Infect Dis. 2004;189(suppl 1):S43-S47.
2601.e8 PART V  Prevention of Infectious Diseases

383. Kaufmann SH. Fact and fiction in tuberculosis vaccine research: 10 years later. confirming the identity of cell substrates and viruses used in vaccine production.
Lancet Infect Dis. 2011;11:633-640. J Virol Methods. 2012;179:201-211.
384. Kayhty H, Karanko V, Peltola H, et al. Serum antibodies to capsular polysaccharide 414. Kuter BJ, Brown ML, Hartzel J, et al. Safety and immunogenicity of a combina-
vaccine of group A Neissera meningitidis followed for three years in infants and tion measles, mumps, rubella and varicella vaccine (ProQuad). Hum Vaccin.
children. J Infect Dis. 1980;142:861-868. 2006;2:205-214.
385. Kelso JM, Jones RT, Yunginger JW. Anaphylaxis to measles, mumps, and rubella 415. Kuter B, Matthews H, Shinefield H, et al. Ten year follow-up of healthy children
vaccine mediated by IgE to gelatin. J Allergy Clin Immunol. 1993;91:867-872. who received one or two injections of varicella vaccine. Pediatr Infect Dis J.
386. Kemp A, Van Asperen P, Mukhi A. Measles immunization in children with clinical 2004;23:132-137.
reactions to egg protein. Am J Dis Child. 1990;144:33-35. 416. Kuter BJ, Weibel RE, Guess HA, et al. Oka/Merck varicella vaccine in healthy
387. Kew OM, Wright PF, Agol VI, et al. Circulating vaccine-derived polioviruses: children: final report of a 2-year efficacy study and 7-year follow-up studies.
current state of knowledge. Bull World Health Organ. 2004;82:16-23. Vaccine. 1991;9:643-647.
388. Keyserling H, Papa T, Koranyi K, et al. Safety, immunogenicity, and immune 417. La Montagne JR, Noble GR, Quinnan GV, et al. Summary of clinical trials of
memory of a novel meningococcal (groups A, C, Y, and W-135) polysaccharide inactivated influenza vaccine—1978. Rev Infect Dis. 1983;5:723-736.
diphtheria toxoid conjugate vaccine (MCV-4) in healthy adolescents. Arch Pediatr 418. LaRussa P, Steinberg S, Meurice F, et al. Transmission of vaccine strain varicella-
Adolesc Med. 2005;159:907-913. zoster virus from a healthy adult with vaccine-associated rash to susceptible
389. Kim-Farley R, Bart S, Stetler H, et al. Clinical mumps vaccine efficacy. Am J household contacts. J Infect Dis. 1997;176:1072-1075.
Epidemiol. 1985;121:593-597. 419. Lavi S, Zimmerman B, Koren G, et al. Administration of measles, mumps and
390. Kim-Farley RJ, Rutherford G, Lichfield P, et al. Outbreak of paralytic poliomyelitis, rubella virus vaccine (live) to egg-allergic children. JAMA. 1990;263:269-271.
Taiwan. Lancet. 1984;2:1322-1324. 420. Le Bouvier GL, Plotkin SA. Precipitin responses to rubella vaccine RA 27-3. J
391. Kimura A, Toneatto D, Kleinschmidt A, et al. Immunogenicity and safety of Infect Dis. 1971;123:220-223.
a multicomponent meningococcal serogroup B vaccine and a quadrivalent 421. LeBaron CW, Bi D, Sullivan BJ, et al. Evaluation of potentially common adverse
meningococcal CRM197 conjugate vaccine against serogroups A, C, W-135, and events associated with the first and second doses of measles-mumps-rubella
Y in adults who are at increased risk for occupational exposure to meningococcal vaccine. Pediatrics. 2006;118:1422-1430.
isolates. Clin Vaccine Immunol. 2011;18:483-486. 422. LeBaron CW, Forghani B, Beck C, et al. Persistence of mumps antibodies after
392. King GE, Hadler SC. Simultaneous administration of childhood vaccines: an 2 doses of measles-mumps-rubella vaccine. J Infect Dis. 2009;199:552-560.
important public health policy that is safe and efficacious. Pediatr Infect Dis J. 423. LeBaron CW, Forghani B, Matter L, et al. Persistence of rubella antibodies after
1994;13:394-407. 2 doses of measles-mumps-rubella vaccine. J Infect Dis. 2009;200:888-899.
393. Klein NP, Bartlett J, Fireman B, et al. Waning Tdap effectiveness in adolescents. 424. Lee SH, Ewert DP, Frederick PD, et al. Resurgence of congenital rubella syndrome
Pediatrics. 2016;137:e20153326. in the 1990s. Report on missed opportunities and failed prevention policies
394. Klein NP, Fireman B, Yih WK, et al. Measles-mumps-rubella-varicella combination among women of childbearing age. JAMA. 1992;267:2616-2620.
vaccine and the risk of febrile seizures. Pediatrics. 2010;126:e1-e8. 425. Lerman SJ, Bollinger M, Brunken JM. Clinical and serological evaluation of
395. Klein NP, Lewis E, Baxter R, et al. Measles-containing vaccines and febrile seizures measles, mumps, and rubella (HPV-77:DE-5 and RA27/3) virus vaccines, singly
in children age 4 to 6 years. Pediatrics. 2012;129:809-814. and in combination. Pediatrics. 1981;68:18-22.
396. Klinge J, Lugauer S, Korn K, et al. Comparison of immunogenicity and reacto- 426. Leroy Z, Broder K, Menschik D, et al. Febrile seizures after 2010-2011 influenza
genicity of a measles, mumps and rubella (MMR) vaccine in German children vaccine in young children, United States: a vaccine safety signal from the vaccine
vaccinated at 9-11, 12-14 or 15-17 months of age. Vaccine. 2000;18:3134-3140. adverse event reporting system. Vaccine. 2012;30:2020-2023.
397. Klugman KP, Gilbertson IT, Koornhof HJ, et al. Protective activity of Vi capsular 427. Leshem E, Moritz RE, Curns AT, et al. Rotavirus vaccines and health care utilization
polysaccharide vaccine against typhoid fever. Lancet. 1987;2(8569):1165-1169. for diarrhea in the United States (2007-2011). Pediatrics. 2014;134:15-23.
398. Klugman KP, Madhi SA, Huebner RE, et al. A trial of a 9-valent pneumococcal 428. Letson GW, Shapiro CN, Kuehn D, et al. Effect of maternal antibody on
conjugate vaccine in children with and those without HIV infection. N Engl J immunogenicity of hepatitis A vaccine in infants. J Pediatr. 2004;144:
Med. 2003;349:1341-1348. 327-332.
399. Kobayashi M, Bennett NM, Gierke R, et al. Intervals Between PCV13 and PPSV23 429. Levine MM. Modern vaccines. Enteric infections. Lancet. 1990;335:958-961.
Vaccines: recommendations of the Advisory Committee on Immunization Practices 430. Levine MM, Noriega F. Vaccines to prevent bacterial enteric infections in children.
(ACIP). MMWR Morb Mortal Wkly Rep. 2015;64:944-947. Pediatr Ann. 1993;22:719-725.
400. Koff RS. Seroepidemiology of hepatitis A in the United States. J Infect Dis. 431. Lewis JE, Chernesky MA, Rawls ML, et al. Epidemic of mumps in a partially
1995;171:S19-S23. immune population. Can Med Assoc J. 1979;121:751-754.
401. Konradsen HB, Pedersen FK, Henrichsen J. Pneumococcal revaccination of 432. Lieberman JM, Chang SJ, Partridge S, et al. Kinetics of maternal hepatitis a
splenectomized children. Pediatr Infect Dis J. 1990;9:258-263. antibody decay in infants: implications for vaccine use. Pediatr Infect Dis J.
402. Koutsky LA, Harper DM. Chapter 13: Current findings from prophylactic HPV 2002;21:347-348.
vaccine trials. Vaccine. 2006;24(suppl 3):S3, 114-121. 433. Lieberman J, Marcy S, Partridge S, et al Hepatitis A vaccine in infants: effect of
403. Kramarz P, Destefano F, Gargiullo PM, et al. Does influenza vaccination prevent maternal antibodies on the antibody response [abstract]. Paper presented at the
asthma exacerbations in children? J Pediatr. 2001;138:306-310. 36th Annual Meeting of the Infectious Disease Society of America, 1998.
404. Kremer JR, Schneider F, Muller CP. Waning antibodies in measles and rubella 434. Lin FY, Ho VA, Khiem HB, et al. The efficacy of a Salmonella typhi Vi conjugate
vaccinees—a longitudinal study. Vaccine. 2006;24:2594-2601. vaccine in two-to-five-year-old children. N Engl J Med. 2001;344:1263-1269.
405. Kretsinger K, Broder KR, Cortese MM, et al. Preventing tetanus, diphtheria, and 435. Lindsey NP, Schroeder BA, Miller ER, et al. Adverse event reports following
pertussis among adults: use of tetanus toxoid, reduced diphtheria toxoid and yellow fever vaccination. Vaccine. 2008;26:6077-6082.
acellular pertussis vaccine recommendations of the Advisory Committee on 436. Loharikar A, Newton AE, Stroika S, et al. Cholera in the United States, 2001-
Immunization Practices (ACIP) and recommendation of ACIP, supported by the 2011: a reflection of patterns of global epidemiology and travel. Epidemiol Infect.
Healthcare Infection Control Practices Advisory Committee (HICPAC), for use of 2015;143:695-703.
Tdap among health-care personnel. MMWR Recomm Rep. 2006;55(RR-17):1-37. 437. Long SS. Toddler-to-mother transmission of varicella-vaccine virus: How bad
406. Kristiansen PA, Jorgensen HJ, Caugant DA. Serogroup A meningococcal conjugate is that? J Pediatrics. 1997;131:10-12.
vaccines in Africa. Expert Rev Vaccines. 2015;14:1441-1458. 438. Lotte A, Wasz-Hockert O, Poisson N, et al. BCG complications: estimates of the
407. Kroger AT, Atkinson WL, Marcuse EK, et al. General recommendations on risks among vaccinated subjects and statistical analysis of their main characteristics.
immunization: recommendations of the Advisory Committee on Immunization Adv Tuberc Res. 1984;21:107-193.
Practices (ACIP). MMWR Recomm Rep. 2006;55(RR-15):1-48. 439. Lowy DR, Schiller JT. Prophylactic human papillomavirus vaccines. J Clin Invest.
408. Kroger A, Sumaya C, Pickering L, et al. General recommendations on immuniza- 2006;116:1167-1173.
tion—recommendations of the Advisory Committee on Immunization Practices 440. Luman ET, Fiore AE, Strine TW, et al. Impact of thimerosal-related changes
(ACIP). MMWR Recomm Rep. 2011;60:1-64. in hepatitis B vaccine birth-dose recommendations on childhood vaccination
409. Kröger L, Korppi M, Brander E, et al. Osteitis caused by bacille Calmette-Guérin coverage. JAMA. 2004;291:2351-2358.
vaccination: a retrospective analysis of 222 cases. J Infect Dis. 1995;172:574-576. 441. MacKellar DA, Valleroy LA, Secura GM, et al. Two decades after vaccine license:
410. Krugman S. Further attenuated measles vaccine: characteristics and use. Rev hepatitis B immunization and infection among young men who have sex with
Infect Dis. 1983;5:477-481. men. Am J Public Health. 2001;91:965-971.
411. Krugman RD, Hardy GE Jr, Sellers C, et al. Antibody persistence after primary 442. MacNeil JR, Bennett N, Farley MM, et al. Epidemiology of infant meningococcal
immunization with trivalent oral poliovirus vaccine. Pediatrics. 1977;60:80-82. disease in the United States, 2006-2012. Pediatrics. 2015;135:e305-e311.
412. Kuehn BM. FDA: benefits of rotavirus vaccination outweigh potential contamina- 443. Macneil JR, Cohn AC, Zell ER, et al. Early estimate of the effectiveness of quad-
tion risk. JAMA. 2010;304:30-31. rivalent meningococcal conjugate vaccine. Pediatr Infect Dis J. 2011;30:451-455.
413. Kumar D, Beach NM, Meng XJ, et al. Use of PCR-based assays for the detection 444. MacNeil JR, Rubin L, Folaranmi T, et al. Use of serogroup B meningococcal
of the adventitious agent porcine circovirus type 1 (PCV1) in vaccines, and for vaccines in adolescents and young adults: recommendations of the Advisory
CHAPTER 245  Active Immunizing Agents 2601.e9

Committee on Immunization Practices, 2015. MMWR Morb Mortal Wkly Rep. 471. McLean HQ, Fiebelkorn AP, Temte JL, et al. Prevention of measles, rubella,
2015;64:1171-1176. congenital rubella syndrome, and mumps, 2013: summary recommendations of
445. MacNeil JR, Rubin LG, Patton M, et al. Recommendations for use of meningococcal the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm
conjugate vaccines in HIV-infected persons—Advisory Committee on Immuniza- Rep. 2013;62(RR-04):1-34.
tion Practices, 2016. MMWR Morb Mortal Wkly Rep. 2016;65:1189-1194. 472. McQuillan GM, Kruszon-Moran D, Deforest A, et al. Serologic immunity to
446. Mahon BE, Newton AE, Mintz ED. Effectiveness of typhoid vaccination in US diphtheria and tetanus in the United States. Ann Intern Med. 2002;136:660-666.
travelers. Vaccine. 2014;32:3577-3579. 473. McVernon J, Andrews N, Slack MP, et al. Risk of vaccine failure after Haemophilus
447. Maldonado YA, Lawrence EC, DeHovitz R, et al. Early loss of passive measles influenzae type b (Hib) combination vaccines with acellular pertussis. Lancet.
antibody in infants of mothers with vaccine-induced immunity. Pediatrics. 2003;361:1521-1523.
1995;96:447-450. 474. Meites E, Kempe A, Markowitz LE. Use of a 2-dose schedule for human papil-
448. Manning SE, Rupprecht CE, Fishbein D, et al. Human rabies prevention—United lomavirus vaccination—updated recommendations of the Advisory Committee
States, 2008: recommendations of the Advisory Committee on Immunization on Immunization Practices. MMWR Morb Mortal Wkly Rep. 2016;65:1405-1408.
Practices. MMWR Recomm Rep. 2008;57(RR-3):1-28. 475. Mellinger AK, Cragan JD, Atkinson WL, et al. High incidence of congenital
449. Maple PA, Efstratiou A, George RC, et al. Diphtheria immunity in UK blood rubella syndrome after a rubella outbreak. Pediatr Infect Dis J. 1995;14:573-578.
donors. Lancet. 1995;345:963-965. 476. Merck & Co. Gardasil (Package Insert). Quadrivalent Human Papillomavirus
450. Marin M, Broder KR, Temte JL, et al. Use of combination measles, mumps, (Types 6, 11, 16, 18) Recombinant Vaccine. Whitehouse Station, NJ: Merck; 2006.
rubella, and varicella vaccine: recommendations of the Advisory Committee 477. Michalik DE, Steinberg SP, Larussa PS, et al. Primary vaccine failure after 1 dose
on Immunization Practices (ACIP). MMWR Recomm Rep. 2010;59(RR-3):1-12. of varicella vaccine in healthy children. J Infect Dis. 2008;197:944-949.
451. Marin M, Guris D, Chaves SS, et al. Prevention of varicella: recommendations of 478. Midthun K, Horne AD, Goldenthal KL. Clinical safety evaluation of combination
the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm vaccines. Dev Biol Stand. 1998;95:245-249.
Rep. 2007;56(RR-4):1-40. 479. Millar EV, O’Brien KL, Watt JP, et al. Effect of community-wide conjugate
452. Marin M, Quinlisk P, Shimabukuro T, et al. Mumps vaccination coverage and pneumococcal vaccine use in infancy on nasopharyngeal carriage through 3
vaccine effectiveness in a large outbreak among college students—Iowa, 2006. years of age: a cross-sectional study in a high-risk population. Clin Infect Dis.
Vaccine. 2008;26:3601-3607. 2006;43:8-15.
453. Marin M, Willis ED, Marko A, et al. Closure of varicella-zoster virus-containing 480. Miller D, Madge N, Diamond J, et al. Pertussis immunisation and serious acute
vaccines pregnancy registry—United States, 2013. MMWR Morb Mortal Wkly neurological illnesses in children. Br Med J. 1993;307:1171-1176.
Rep. 2014;63:732-733. 481. Miller D, Wadsworth J, Diamond J, et al. Pertussis vaccine and whooping cough
454. Markowitz L, Albrecht P, Orenstein W, et al. Persistence of measles antibody as risk factors in acute neurologic illness and death in young children. Dev Biol
after revaccination. J Infect Dis. 1992;166:205-208. Stand. 1985;61:389-394.
455. Markowitz LE, Albrecht P, Rhodes P, et al. Changing levels of measles antibody titers 482. Mofenson LM, Brady MT, Danner SP, et al. Guidelines for the prevention and
in women and children in the United States: Impact on response to vaccination. treatment of opportunistic infections among HIV-exposed and HIV-infected
Pediatrics. 1996;97:53-58. children: recommendations from CDC, the National Institutes of Health, the
456. Markowitz LE, Dunne EF, Saraiya M, et al. Quadrivalent human papillomavirus HIV Medicine Association of the Infectious Diseases Society of America, the
vaccine: recommendations of the Advisory Committee on Immunization Practices Pediatric Infectious Diseases Society, and the American Academy of Pediatrics.
(ACIP). MMWR Recomm Rep. 2007;56(RR-2):1-24. MMWR Recomm Rep. 2009;58(RR-11):1-166.
457. Markowitz LE, Dunne EF, Saraiya M, et al. Human papillomavirus vaccination: 483. Molano M, Van den Brule A, Plummer M, et al. Determinants of clearance of
recommendations of the Advisory Committee on Immunization Practices (ACIP). human papillomavirus infections in Colombian women with normal cytol-
MMWR Recomm Rep. 2014;63(RR-05):1-30. ogy: a population-based, 5-year follow-up study. Am J Epidemiol. 2003;158:
458. Markowitz LE, Preblud SR, Fine PE, et al. Duration of live measles vaccine-induced 486-494.
immunity. Pediatr Infect Dis J. 1990;9:101-110. 484. Monath T, Cetron M, Teuwen D. Yellow fever vaccine. In: Plotkin S, Orenstein
459. Markowitz LE, Preblud SR, Orenstein WA, et al. Patterns of transmission in W, Offit P, eds. Vaccines. 5th ed. Philadelphia: Saunders-Elsevier; 2008:959-1055.
measles outbreaks in the United States, 1985-1986. N Engl J Med. 1989;320:75-81. 485. Monroe BP, Yager P, Blanton J, et al. Rabies surveillance in the United States
460. Marshall GS, Marchant CD, Blatter M, et al. Co-administration of a novel during 2014. J Am Vet Med Assoc. 2016;248:777-788.
Haemophilus influenzae type b and Neisseria meningitidis serogroups C and 486. Monto AS, Kioumehr F. The Tecumseh Study of Respiratory Illness. IX. Occurence
Y-tetanus toxoid conjugate vaccine does not interfere with the immune response of influenza in the community, 1966—1971. Am J Epidemiol. 1975;102:553-563.
to antigens contained in infant vaccines routinely used in the United States. 487. Moore MR, Link-Gelles R, Schaffner W, et al. Effect of use of 13-valent pneumococ-
Hum Vaccin. 2011;7:258-264. cal conjugate vaccine in children on invasive pneumococcal disease in children
461. Marshall HS, Richmond PC, Nissen MD, et al. A phase 2 open-label safety and and adults in the USA: analysis of multisite, population-based surveillance. Lancet
immunogenicity study of a meningococcal B bivalent rLP2086 vaccine in healthy Infect Dis. 2015;15:301-309.
adults. Vaccine. 2013;31:1569-1575. 488. Morgan JL, Baggari SR, McIntire DD, et al. Pregnancy outcomes after antepar-
462. Martin M, Tsai T, Cropp B, et al. Fever and multi-system organ failure associ- tum tetanus, diphtheria, and acellular pertussis vaccination. Obstet Gynecol.
ated with 17D-204 yellow fever vaccination: a report of four cases. Lancet. 2015;125:1433-1438.
2001;358:98-104. 489. Moro PL, Cragan J, Tepper N, et al. Enhanced surveillance of tetanus toxoid,
463. Martinon-Torres F, Czajka H, Center KJ, et al. 13-valent pneumococcal conjugate reduced diphtheria toxoid, and acellular pertussis (Tdap) vaccines in pregnancy
vaccine (PCV13) in preterm versus term infants. Pediatrics. 2015;135:e876-e886. in the Vaccine Adverse Event Reporting System (VAERS), 2011-2015. Vaccine.
464. Mast EE, Margolis HS, Fiore AE, et al. A comprehensive immunization strategy 2016;34:2349-2353.
to eliminate transmission of hepatitis B virus infection in the United States: 490. Mortimer EA Jr. Pertussis and pertussis vaccine. In: Aronoff SC, ed. Advances
recommendations of the Advisory Committee on Immunization Practices (ACIP) in Pediatric Infectious Diseases. Vol. 5. Chicago: Year Book Medical Publisher;
part 1: immunization of infants, children, and adolescents. MMWR Recomm 1990:1-27.
Rep. 2005;54(RR-16):1-31. 491. Moxon ER, Rappuoli R. Haemophilus influenzae infections and whooping cough.
465. Mast EE, Weinbaum CM, Fiore AE, et al. A comprehensive immunization strategy Lancet. 1990;335:1324-1329.
to eliminate transmission of hepatitis B virus infection in the United States: 492. Munoz N, Bosch FX, de Sanjose S, et al. Epidemiologic classification of
recommendations of the Advisory Committee on Immunization Practices (ACIP). human papillomavirus types associated with cervical cancer. N Engl J Med.
Part II: Immunization of adults. MMWR Recomm Rep. 2006;55(RR-16):1-33, 2003;348:518-527.
quiz CE31-34. 493. Murphy TV. Haemophilus influenzae vaccines: 1997. Adv Pediatr Infect Dis.
466. Mathias RG, Meekison WG, Arcand TA, et al. The role of secondary vaccine 1997;13:279-304.
failures in measles outbreaks. Am J Public Health. 1989;79:475-478. 494. Murphy TV, Denniston MM, Hill HA, et al. Progress toward eliminating hepatitis
467. Mazurek GH, Jereb J, Vernon A, et al. Updated guidelines for using interferon A disease in the United States. MMWR Suppl. 2016;65:29-41.
gamma release assays to detect Mycobacterium tuberculosis infection—United 495. Murphy TV, Gargiullo PM, Massoudi MS, et al. Intussusception among infants
States, 2010. MMWR Recomm Rep. 2010;59(RR-5):1-25. given an oral rotavirus vaccine. N Engl J Med. 2001;344:564-572.
468. McBean AM, Thoms ML, Albrecht P, et al. Serologic response to oral polio 496. Myaux JA, Unicomb L, Besser RE, et al. Effect of diarrhea on the humoral response
vaccine and enhanced-potency inactivated polio vaccine. Am J Epidemiol. to oral polio vaccination. Pediatr Infect Dis J. 1996;15:204-209.
1988;128:615-628. 497. National Institute of Allergy and Infectious Diseases. The Jordan Report—Acceler-
469. McClenahan SD, Krause PR, Uhlenhaut C. Molecular and infectivity studies of ated Development of Vaccines, 2012. Washington, DC: National Institutes of Health;
porcine circovirus in vaccines. Vaccine. 2011;29:4745-4753. 2012.
470. McLaughlin M, Thomas P, Onorato I, et al. Live virus vaccines in human 498. National Vaccine Advisory Committee. Standards for child and adolescent
immunodeficiency virus-infected children: a retrospective survey. Pediatrics. immunization practices. National Vaccine Advisory Committee. Pediatrics.
1988;82:229-233. 2003;112:958-963.
2601.e10 PART V  Prevention of Infectious Diseases

499. Negri E, Colombo C, Giordano L, et al. Influenza vaccine in healthy children: 528. Payne AB, Link-Gelles R, Azonobi I, et al. Invasive pneumococcal disease among
a meta-analysis. Vaccine. 2005;23:2851-2861. children with and without sickle cell disease in the United States, 1998 to 2009.
500. Nelson GE, Aguon A, Valencia E, et al. Epidemiology of a mumps outbreak in a Pediatr Infect Dis J. 2013;32:1308-1312.
highly vaccinated island population and use of a third dose of measles-mumps- 529. Peebles TC, Levine L, Eldred MC, et al. Tetanus-toxoid emergency boosters: a
rubella vaccine for outbreak control—Guam 2009 to 2010. Pediatr Infect Dis J. reappraisal. N Engl J Med. 1969;280:575-581.
2013;32:374-380. 530. Peltola H, Heinonen O. Frequency of true adverse reactions to measles-mumps-
501. Neuzil KM, Dupont WD, Wright PF, et al. Efficacy of inactivated and cold-adapted rubella vaccine. Lancet. 1986;1:939-942.
vaccines against influenza A infection, 1985 to 1990: the pediatric experience. 531. Peltola H, Makela H, Kayhty H, et al. Clinical efficacy of meningococcus group
Pediatr Infect Dis J. 2001;20:733-740. A capsular polysaccharide vaccine in children three months to five years of age.
502. Neuzil KM, Jackson LA, Nelson J, et al. Immunogenicity and reactogenicity N Engl J Med. 1977;297:686-691.
of 1 versus 2 doses of trivalent inactivated influenza vaccine in vaccine-naive 532. Peter G. Vaccine crisis: an emerging societal problem. J Infect Dis. 1985;151:981-983.
5-8-year-old children. J Infect Dis. 2006;194:1032-1039. 533. Petrosky E, Bocchini JA Jr, Hariri S, et al. Use of 9-valent human papillomavirus
503. Newton A, Routh J, Barbara E, Mahon B. Typhoid and paratyphoid fever. In: (HPV) vaccine: updated HPV vaccination recommendations of the advisory
Brunette G, ed. CDC Health Information for International Travel, 2016. New committee on immunization practices. MMWR Morb Mortal Wkly Rep.
York: Oxford University Press; 2016. 2015;64:300-304.
504. Nguyen HQ, Jumaan AO, Seward JF. Decline in mortality due to varicella after 534. Phadke VK, Bednarczyk RA, Salmon DA, et al. Association between vaccine
implementation of varicella vaccination in the United States. N Engl J Med. refusal and vaccine-preventable diseases in the United States: a review of measles
2005;352:450-458. and pertussis. JAMA. 2016;315:1149-1158.
505. Nkowane BM, Bart KJ, Orenstein WA, et al. Measles outbreak in a vaccinated 535. Phua KB, Quak SH, Lee BW, et al. Evaluation of RIX4414, a live, attenuated
school population. Epidemiology, strains of transmission and the role of vaccine rotavirus vaccine, in a randomized, double-blind, placebo-controlled phase 2
failures. Am J Public Health. 1987;77:434-438. trial involving 2464 Singaporean infants. J Infect Dis. 2005;192(suppl 1):S6-S16.
506. Nolan T, McVernon J, Skeljo M, et al. Immunogenicity of a monovalent 2009 536. Piazza M, Safary A, Vegnente A, et al. Safety and immunogenicity of hepatitis
influenza A(H1N1) vaccine in infants and children: a randomized trial. JAMA. A vaccine in infants: a candidate for inclusion in the childhood vaccination
2010;303:37-46. programme. Vaccine. 1999;17:585-588.
507. Nolan T, Richmond P, Marshall H, et al. Immunogenicity and safety of an 537. Pichichero ME, Edwards KM, Anderson EL, et al. Safety and immunogenicity of
investigational combined Haemophilus influenzae type b–Neisseria meningitidis six acellular pertussis vaccines and one whole-cell vaccine given as a fifth dose
serogroups C and Y–tetanus toxoid conjugate vaccine. Pediatr Infect Dis J. in four- to six-year-old children. Pediatrics. 2000;105:e11.
2011;30:190-196. 538. Plennevaux E, Sheldon E, Blatter M, et al. Immune response after a single vac-
508. Noronha AS, Markowitz LE, Dunne EF. Systematic review of human papillomavirus cination against 2009 influenza A H1N1 in USA: a preliminary report of two
vaccine coadministration. Vaccine. 2014;32:2670-2674. randomised controlled phase 2 trials. Lancet. 2010;375:41-48.
509. Nuorti JP, Whitney CG. Prevention of pneumococcal isease among infants and 539. Plotkin SA. Increasing complexity of vaccine development. J Infect Dis.
children—use of 13-valent pneumococcal conjugate vaccine and 23-valent 2015;212(suppl 1):S12-S16.
pneumococcal polysaccharide: vaccine recommendations of the Advisory 540. Plotkin SA, Buser F. History of RA27/3 rubella vaccine. Rev Infect Dis. 1985;7(suppl
Committee on Immunization Practices (ACIP). Morbidity and Mortality Weekly 1):S77-S78.
Report. 2010;59(RR-11):1-19. 541. Plotkin S, Cadoz M. The acellular pertussis vaccine trials: an interpretation.
510. O’Brien K, Ruff A, Louis M, et al. Bacillus Calmette-Guérin complications in Pediatr Infect Dis J. 1997;16:508-517.
children born to HIV-1-infected women with a review of the literature. Pediatrics. 542. Poehling KA, Edwards KM, Weinberg GA, et al. The underrecognized burden
1995;95:414-418. of influenza in young children. N Engl J Med. 2006;355:31-40.
511. Ong BA, Forester J, Fallot A. Does influenza vaccination improve pediatric asthma 543. Poehling KA, Talbot TR, Griffin MR, et al. Invasive pneumococcal disease among
outcomes? J Asthma. 2009;46:477-480. infants before and after introduction of pneumococcal conjugate vaccine. JAMA.
512. Onorato I, Markowitz L, Oxtoby M. Childhood immunization, vaccine-preventable 2006;295:1668-1674.
diseases and infection with human immunodeficiency virus. Pediatr Infect Dis 544. Popow-Kraupp T, Kundi M, Ambrosch F, et al. A controlled trial for evaluating
J. 1988;6:588-595. two live attenuated mumps-measles vaccines (Urabe Am 9-Schwarz and Jeryl
513. Onorato IM, Wassilak SG, Meade B. Efficacy of whole-cell pertussis vaccine in Lynn-Moraten) in young children. J Med Virol. 1986;18:69-79.
preschool children in the United States. JAMA. 1992;267:2745-2749. 545. Preblud SR. Some current issues relating to rubella vaccine. JAMA.
514. Orenstein WA, Albrecht P, Herrmann KL, et al. The plaque-neutralization 1985;254:253-256.
test as a maesure of prior exposure to measles virus. J Infect Dis. 1987;155: 546. Read RC, Baxter D, Chadwick DR, et al. Effect of a quadrivalent meningococcal
146-149. ACWY glycoconjugate or a serogroup B meningococcal vaccine on meningo-
515. Orenstein WA, Bart KJ, Hinman HR, et al. The opportunity and obligation to coccal carriage: an observer-blind, phase 3 randomised clinical trial. Lancet.
eliminate rubella from the United States. JAMA. 1984;251:1988-1994. 2014;384:2123-2131.
516. Organization WH. Global polio eradication initiative. www.polioeradication.org. 547. Read TR, Hocking JS, Chen MY, et al. The near disappearance of genital warts
517. Oya A. Japanese encephalitis vaccine. Acta Pediatr Jpn. 1988;30:175-184. in young women 4 years after commencing a national human papillomavirus
518. Ozanne G, d’Halewyn M. Secondary immune response in a vaccinated population (HPV) vaccination programme. Sex Transm Infect. 2011;87:544-547.
during a large measles epidemic. Clin Microbiol. 1992;30:1778-1782. 548. Redd SC, King GE, Heath JL, et al. Comparison of vaccination with measles-
519. Palumbo P, Hoyt L, Demasio K, et al. Population-based study of measles and mumps-rubella vaccine at 9, 12, and 15 months of age. J Infect Dis. 2004;189(suppl
measles immunization in human immunodeficiency virus-infected children. 1):S116-S122.
Pediatr Infect Dis J. 1992;11:1008-1014. 549. Reef SE, Cochi SL. The evidence for the elimination of rubella and congenital
520. Papania MJ, Wallace GS, Rota PA, et al. Elimination of endemic measles, rubella, rubella syndrome in the United States: a public health achievement. Clin Infect
and congenital rubella syndrome from the Western hemisphere: the US experience. Dis. 2006;43(suppl 3):S123-S125.
JAMA Pediatr. 2014;168:148-155. 550. Reef SE, Redd SB, Abernathy E, et al. The epidemiological profile of rubella
521. Parashar UD, Gibson CJ, Bresse JS, et al. Rotavirus and severe childhood diarrhea. and congenital rubella syndrome in the United States, 1998-2004: the evidence
Emerg Infect Dis. 2006;12:304-306. for absence of endemic transmission. Clin Infect Dis. 2006;43(suppl 3):
522. Park CL, Frank AL, Sullivan M, et al. Influenza vaccination of children during S126-S132.
acute asthma exacerbation and concurrent prednisone therapy. Pediatrics. 551. Reef SE, Redd SB, Abernathy E, et al. Evidence used to support the achievement
1996;98:196-200. and maintenance of elimination of rubella and congenital rubella syndrome in
523. Parker Fiebelkorn A, Redd SB, Gallagher K, et al. Measles in the United States the United States. J Infect Dis. 2011;204(suppl 2):S593-S597.
during the postelimination era. J Infect Dis. 2010;202:1520-1528. 552. Reefhuis J, Honein MA, Whitney CG, et al. Risk of bacterial meningitis in children
524. Patel MM, Lopez-Collada VR, Bulhoes MM, et al. Intussusception risk and with cochlear implants. N Engl J Med. 2003;349:435-445.
health benefits of rotavirus vaccination in Mexico and Brazil. N Engl J Med. 553. Reingold AL, Broome CV, Hightower AW, et al. Age-specific differences in duration
2011;364:2283-2292. of clinical protection after vaccination with meningococcal polysaccharide A
525. Patja A, Davidkin I, Kurki T, et al. Serious adverse events after measles-mumps- vaccine. Lancet. 1985;2(8447):114-118.
rubella vaccination during a fourteen-year prospective follow-up. Pediatr Infect 554. Rennels MB, Deloria MA, Pichichero ME, et al. Extensive swelling after booster
Dis J. 2000;19:1127-1134. doses of acellular pertussis-tetanus-diphtheria vaccines. Pediatrics. 2000;105:e12.
526. Patriarca PA, Wright PF, John TJ. Factors affecting the immunogenicity of oral 555. Richard JL, Zwahlen M, Feuz M, et al. Comparison of the effectiveness of two
poliovirus vaccine in developing countries: review. Rev Infect Dis. 1991;13:926-939. mumps vaccines during an outbreak in Switzerland in 1999 and 2000: a case-cohort
527. Payne DC, Edwards KM, Bowen MD, et al. Sibling transmission of vaccine- study. Eur J Epidemiol. 2003;18:569-577.
derived rotavirus (RotaTeq) associated with rotavirus gastroenteritis. Pediatrics. 556. Richmond PC, Marshall HS, Nissen MD, et al. Safety, immunogenicity, and
2010;125:e438-e441. tolerability of meningococcal serogroup B bivalent recombinant lipoprotein 2086
CHAPTER 245  Active Immunizing Agents 2601.e11

vaccine in healthy adolescents: a randomised, single-blind, placebo-controlled, 584. Schlegel M, Osterwalder JJ, Galeazzi RL, et al. Comparative efficacy of three
phase 2 trial. Lancet Infect Dis. 2012;12:597-607. mumps vaccines during disease outbreak in Eastern Switzerland: cohort study.
557. Rigau-Perez JG, Overturf GD, Chan LS, et al. Reactions to booster pneumococcal BMJ. 1999;319:352.
vaccination in patients with sickle cell disease. J Pediatr Infect Dis. 1983;2:199-202. 585. Schmitt-Grohe S, Stehr K, Cherry JD, et al. Minor adverse events in a comparative
558. Ritzwoller DP, Bridges CB, Shetterly S, et al. Effectiveness of the 2003-2004 efficacy trial in Germany in infants receiving either the Lederle/Takeda acellular
influenza vaccine among children 6 months to 8 years of age, with 1 vs 2 doses. pertussis component DTP (DTaP) vaccine, the Lederle whole-cell component
Pediatrics. 2005;116:153-159. DTP (DTP) or DT vaccine. The Pertussis Vaccine Study Group. Dev Biol Stand.
559. Rivera L, Pena LM, Stainier I, et al. Horizontal transmission of a human rotavirus 1997;89:113-118.
vaccine strain—a randomized, placebo-controlled study in twins. Vaccine. 586. Schuchat A, Robinson K, Wenger JD, et al. Bacterial meningitis in the United
2011;29:9508-9513. States in 1995. Active Surveillance Team. N Engl J Med. 1997;337:970-976.
560. Robinson CL, Advisory Committee on Immunization Practices (ACIP), ACIP 587. Schuller E, Klingler A, Dubischar-Kastner K, et al. Safety profile of the Vero
Child/Adolescent Immunization Work Group. Advisory Committee on Immuniza- cell-derived Japanese encephalitis virus (JEV) vaccine IXIARO(®). Vaccine.
tion Practices recommended immunization schedules for persons aged 0 through 2011;29:8669-8676.
18 years—United States, 2016. MMWR Morb Mortal Wkly Rep. 2016;65:86-87. 588. Schwarz AJ, Jackson JE, Ehrenkranz NJ, et al. Clinical evaluation of a new
561. Robinson J, Lemay M, Vaudry WL. Congenital rubella after anticipated mater- measles-mumps-rubella trivalent vaccine. Am J Dis Child. 1975;129:1408-1412.
nal immunity: two cases and a review of the literature. Pediatr Infect Dis J. 589. Schwarzer S, Reibel S, Lang AB, et al. Safety and characterization of the immune
1994;13:812-815. response engendered by two combined measles, mumps and rubella vaccines.
562. Rodrigues LC, Diwan VK, Wheeler JG. Protective effect of BCG against tuber- Vaccine. 1998;16:298-304.
culous meningitis and miliary tuberculosis: a meta-analysis. Int J Epidemiol. 590. Seager C, Moriarity J, Ngai A, et al. Low incidence of adverse experiences after
1993;22:1154-1158. measles or measles-rubella mass revaccination at a college campus. Vaccine.
563. Rodriguez ZM, Goveia MG, Stek JE, et al. Concomitant use of an oral live 1994;12:1018-1020.
pentavalent human-bovine reassortant rotavirus vaccine with licensed parenteral 591. Senders S, Bhuyan P, Jiang Q, et al. Immunogenicity, tolerability and safety
pediatric vaccines in the United States. Pediatr Infect Dis J. 2007;26:221-227. in adolescents of bivalent rLP2086, a meningococcal serogroup B vaccine,
564. Rogers DJ, Wilson AJ, Hay SI, et al. The global distribution of yellow fever and coadministered with quadrivalent human papilloma virus vaccine. Pediatr Infect
dengue. Adv Parasitol. 2006;62:181-220. Dis J. 2016;35:548-554.
565. Ross SC, Densen P. Complement deficiency states and infection: epidemiology, 592. Seward JF, Marin M, Vazquez M. Varicella vaccine effectiveness in the US vac-
pathogenesis and consequences of neisserial and other infections in an immune cination program: a review. J Infect Dis. 2008;197(suppl 2):S82-S89.
deficiency. Medicine (Baltimore). 1984;63:243-273. 593. Seward JF, Zhang JX, Maupin TJ, et al. Contagiousness of varicella in vaccinated
566. Routh J, Newton A, Mintz E. Cholera. In: Brunette G, ed. CDC Health Information cases: a household contact study. JAMA. 2004;292:704-708.
for International Travel, 2016. New York: Oxford University Press; 2016. 594. Shapiro ED, Berg AT, Austrian R, et al. The protective efficacy of polyvalent
567. Ruben FL, Hankins WA, Zeigler Z, et al. Antibody responses to meningococcal pneumococcal polysaccharide vaccine. N Engl J Med. 1991;325:1453-1460.
polysaccharide vaccine in adults without a spleen. Am J Med. 1984;76:115-121. 595. Shapiro ED, Vazquez M, Esposito D, et al. Effectiveness of 2 doses of varicella
568. Rubin LG, Levin MJ, Ljungman P, et al. 2013 IDSA clinical practice guideline for vaccine in children. J Infect Dis. 2011;203:312-315.
vaccination of the immunocompromised host. Clin Infect Dis. 2014;58:309-318. 596. Sharrar R, LaRussa P, Galea S, et al. The postmarketing safety profile of varicella
569. Ruiz-Palacios GM, Perez-Schael I, Velazquez FR, et al. Safety and efficacy of vaccine. Vaccine. 2000;19:916-923.
an attenuated vaccine against severe rotavirus gastroenteritis. N Engl J Med. 597. Sheldon EA, Schwartz H, Jiang Q, et al. A phase 1, randomized, open-label,
2006;354:11-22. active-controlled trial to assess the safety of a meningococcal serogroup B bivalent
570. Rupprecht CE, Briggs D, Brown CM, et al. Use of a reduced (4-dose) vaccine rLP2086 vaccine in healthy adults. Hum Vaccin Immunother. 2012;8:888-895.
schedule for postexposure prophylaxis to prevent human rabies: recommendations 598. Shill M, Baynes RD, Miller SD. Fatal rabies encephalitis despite appropriate
of the advisory committee on immunization practices. MMWR Recomm Rep. post-exposure prophylaxis: a case report. N Engl J Med. 1987;316:1257-1258.
2010;59(RR-2):1-9. 599. Shin S, Desai SN, Sah BK, et al. Oral vaccines against cholera. Clin Infect Dis.
571. Safadi MA, Cintra OA. Epidemiology of meningococcal disease in Latin America: 2011;52:1343-1349.
current situation and opportunities for prevention. Neurol Res. 2010;32: 600. Shinefield HR, Black S. Efficacy of pneumococcal conjugate vaccines in large
263-271. scale field trials. Pediatr Infect Dis J. 2000;19:394-397.
572. Saha A, Chowdhury MI, Khanam F, et al. Safety and immunogenicity study of a 601. Shui IM, Baggs J, Patel M, et al. Risk of intussusception following administration
killed bivalent (O1 and O139) whole-cell oral cholera vaccine Shanchol, in Ban- of a pentavalent rotavirus vaccine in US infants. JAMA. 2012;307:598-604.
gladeshi adults and children as young as 1 year of age. Vaccine. 2011;29:8285-8292. 602. Shuler CM, Iwamoto M, Bridges CB, et al. Vaccine effectiveness against medically
573. Salzman M, Garcia C. Postexposure varicella vaccination in siblings of children attended, laboratory-confirmed influenza among children aged 6 to 59 months,
with active varicella. Pediatr Infect Dis J. 1998;17:256-257. 2003-2004. Pediatrics. 2007;119:e587-e595.
574. Salzman MB, Sharrar RG, Steinberg S, et al. Transmission of varicella-vaccine 603. Siber GR, Werner BG, Halsey NA, et al. Interference of immune globulin with
virus from a healthy 12-month-old child to his pregnant mother. J Pediatr. measles and rubella immunization. J Pediatr. 1993;122:204-211.
1997;131:151-154. 604. Siddiqui FJ, Rabbani F, Hasan R, et al. Typhoid fever in children: some epi-
575. Santolaya ME, O’Ryan ML, Valenzuela MT, et al. Immunogenicity and tolerability demiological considerations from Karachi, Pakistan. Int J Infect Dis. 2006;10:
of a multicomponent meningococcal serogroup B (4CMenB) vaccine in healthy 215-222.
adolescents in Chile: a phase 2b/3 randomised, observer-blind, placebo-controlled 605. Simanjuntak CH, Paleologo FP, Punjabi NH, et al. Oral immunisation against
study. Lancet. 2012;379:617-624. typhoid fever in Indonesia with Ty21a vaccine. Lancet. 1991;338:1055-1059.
576. Santolaya ME, O’Ryan M, Valenzuela MT, et al. Persistence of antibodies in 606. Simoes EA, John TJ. The antibody response of seronegative infants to inactivated
adolescents 18-24 months after immunization with one, two, or three doses poliovirus vaccine of enhanced potency. J Biol Stand. 1986;14:127-131.
of 4CMenB meningococcal serogroup B vaccine. Hum Vaccin Immunother. 607. Singh R, John TJ, Cherian T, et al. Immune response to measles, mumps &
2013;9:2304-2310. rubella vaccine at 9, 12 & 15 months of age. Indian J Med Res. 1994;100:155-159.
577. Santosham M, Wolff M, Reid R, et al. The efficacy in Navajo infants of a conjugate 608. Singleton R, Hammitt L, Hennessy T, et al. The Alaska Haemophilus influenzae
vaccine consisting of Haemophilus influenzae type b polysaccharide and Neisseria type b experience: lessons in controlling a vaccine-preventable disease. Pediatrics.
meningitidis outer-membrane protein complex. N Engl J Med. 1991;324:1767-1772. 2006;118:e421-e429.
578. Sartorius B, Penttinen P, Nilsson J, et al. An outbreak of mumps in Sweden, 609. Sinha A, Sazawal S, Kumar R, et al. Typhoid fever in children aged less than 5
February-April 2004. Euro Surveill. 2005;10:191-193. years. Lancet. 1999;354:734-737.
579. Satterwhite CL, Torrone E, Meites E, et al. Sexually transmitted infections among 610. Smith PJ, Marcuse EK, Seward JF, et al. Children and adolescents unvaccinated
US women and men: prevalence and incidence estimates, 2008. Sex Transm Dis. against measles: geographic clustering, parents’ beliefs, and missed opportunities.
2013;40:187-193. Public Health Rep. 2015;130:485-504.
580. Schaffner W, Fleet WF, Kilroy AW, et al. Polyneuropathy following rubella 611. Smith CK, McNeal MM, Meyer NR, et al. Rotavirus shedding in premature
immunization. A follow-up study and review of the problem. Am J Dis Child. infants following first immunization. Vaccine. 2011;29:8141-8146.
1974;127:684-688. 612. Snijders BE, van Lier A, van de Kassteele J, et al. Mumps vaccine effective-
581. Schaffzin JK, Pollock L, Schulte C, et al. Effectiveness of previous mumps vac- ness in primary schools and households, the Netherlands, 2008. Vaccine.
cination during a summer camp outbreak. Pediatrics. 2007;120:e862-e868. 2012;30:2999-3002.
582. Schillie S, Murphy TV, Fenlon N, et al. Update: shortened interval for postvac- 613. Song J-H. The relationship between pneumococcal serotypes and antibiotic
cination serologic testing of infants born to hepatitis B-infected mothers. MMWR resistance. Vaccine. 2012;30:2728.
Morb Mortal Wkly Rep. 2015;64:1118-1120. 614. Sotir MJ, Esposito DH, Barnett ED, et al. Measles in the 21st century, a continuing
583. Schillie S, Walker T, Veselsky S, et al. Outcomes of infants born to women infected preventable risk to travelers: data from the GeoSentinel Global Network. Clin
with hepatitis B. Pediatrics. 2015;135:e1141-e1147. Infect Dis. 2016;62:210-212.
2601.e12 PART V  Prevention of Infectious Diseases

615. Sprauer MA, Markowitz LE, Nicholson JK, et al. Response of human immuno- 643. Troisi CL, Hollinger FB, Krause DS, et al. Immunization of seronegative infants
deficiency virus-infected adults to measles- rubella vaccination. J Acquir Immune with hepatitis A vaccine (HAVRIX; SKB): a comparative study of two dosing
Defic Syndr. 1993;6:1013-1016. schedules. Vaccine. 1997;15:1613-1617.
616. Stanley M, Lowy DR, Frazer I. Chapter 12: prophylactic HPV vaccines: underlying 644. Trollfors B, Taranger J, Lagergard T, et al. A placebo-controlled trial of a pertussis-
mechanisms. Vaccine. 2006;24(suppl 3):106-113. toxoid vaccine. N Engl J Med. 1995;333:1045-1050.
617. Staples JE, Bocchini JA Jr, Rubin L, et al. Yellow fever vaccine booster doses: 645. Tse A, Tseng HF, Greene SK, et al. Signal identification and evaluation for risk
recommendations of the Advisory Committee on Immunization Practices, 2015. of febrile seizures in children following trivalent inactivated influenza vaccine
MMWR Morb Mortal Wkly Rep. 2015;64:647-650. in the Vaccine Safety Datalink Project, 2010-2011. Vaccine. 2012;30:2024-2031.
618. Staples JE, Gershman M, Fischer M. Yellow fever vaccine: recommendations of 646. Tugwell BD, Lee LE, Gillette H, et al. Chickenpox outbreak in a highly vaccinated
the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm school population. Pediatrics. 2004;113:455-459.
Rep. 2010;59(RR-7):1-27. 647. Usonis V, Bakasenas V, Chitour K, et al. Comparative study of reactogenicity and
619. Steffen R, Kane MA, Shapiro CN, et al. Epidemiology and prevention of hepatitis immunogenicity of new and established measles, mumps and rubella vaccines
A in travelers. JAMA. 1994;272:885-889. in healthy children. Infection. 1998;26:222-226.
620. Stokley S, Jeyarajah J, Yankey D, et al. Human papillomavirus vaccination coverage 648. Usonis V, Bakasenas V, Kaufhold A, et al. Reactogenicity and immunogenicity of
among adolescents, 2007-2013, and postlicensure vaccine safety monitoring, a new live attenuated combined measles, mumps and rubella vaccine in healthy
2006-2014—United States. MMWR Morb Mortal Wkly Rep. 2014;63:620-624. children. Pediatr Infect Dis J. 1999;18:42-48.
621. Stone M, Vannier A, Storch S, et al. Brief report: meningitis due to iatrogenic BCG 649. Vadheim CM, Greenberg DP, Partridge S, et al. Effectiveness and safety of an
infection in two immunocompromised children. N Engl J Med. 1995;333:561-563. Haemophilus influenzae type b conjugate vaccine (PRP-T) in young infants.
622. Stratton KR, Howe CJ, Johnston RB Jr. Adverse events associated with childhood Kaiser-UCLA Vaccine Study Group. Pediatrics. 1993;92:272-279.
vaccines other than pertussis and rubella. Summary of a report from the Institute 650. Van Damme P, Banatvala J, Fay O, et al. Hepatitis A booster vaccination: is there
of Medicine. JAMA. 1994;271:1602-1605. a need? Lancet. 2003;362:1065-1071.
623. Sugaya N, Nerome K, Ishida M, et al. Efficacy of inactivated vaccine in pre- 651. Van Effelterre TP, Zink TK, Hoet BJ, et al. A mathematical model of hepatitis
venting antigenically drifted influenza type A and well-matched type B. JAMA. a transmission in the United States indicates value of universal childhood
1994;272:1122-1126. immunization. Clin Infect Dis. 2006;43:158-164.
624. Sugg WC, Finger JA, Levine RH, et al. Field evaluation of live virus mumps 652. Van Herck K, Van Damme P. Inactivated hepatitis A vaccine-induced antibodies:
vaccine. J Pediatr. 1968;72:461-466. follow-up and estimates of long-term persistence. J Med Virol. 2001;63:1-7.
625. Sukumaran L, McCarthy NL, Kharbanda EO, et al. Association of Tdap vaccination 653. Vandermeulen C, Clement F, Roelants M, et al. Evaluation of cellular immunity
with acute events and adverse birth outcomes among pregnant women with to mumps in vaccinated individuals with or without circulating antibodies up
prior tetanus-containing immunizations. JAMA. 2015;314:1581-1587. to 16 years after their last vaccination. J Infect Dis. 2009;199:1457-1460.
626. Sullivan KM, Halpin TJ, Marks JS, et al. Effectiveness of mumps vaccine in a 654. Vasquez M, LaRussa P, Gershon A, et al. The effectiveness of varicella vaccine
school outbreak. Am J Dis Child. 1985;139:909-912. in clinical practice. N Engl J Med. 2001;344:1007-1009.
627. Sun Y, Christensen J, Hviid A, et al. Risk of febrile seizures and epilepsy after 655. Velázquez FR, Colindres RE, Grajales C, et al. Postmarketing surveillance of
vaccination with diphtheria, tetanus, acellular pertussis, inactivated poliovirus, intussusception following mass introduction of the attenuated human rotavirus
and Haemophilus influenzae type B. JAMA. 2012;307:823-831. vaccine in Mexico. Pediatr Infect Dis J. 2012;31:736-44 710. 1097/INF.1090b101
628. Sur D, Kanungo S, Sah B, et al. Efficacy of a low-cost, inactivated whole-cell oral 3e318253add318253.
cholera vaccine: results from 3 years of follow-up of a randomized, controlled 656. Verstraeten T, Jumaan AO, Mullooly JP, et al. A retrospective cohort study of the
trial. PLoS Negl Trop Dis. 2011;5:e1289. association of varicella vaccine failure with asthma, steroid use, age at vaccination,
629. Sur D, Ochiai RL, Bhattacharya SK, et al. A cluster-randomized effectiveness and measles-mumps-rubella vaccination. Pediatrics. 2003;112:e98-e103.
trial of Vi typhoid vaccine in India. N Engl J Med. 2009;361:335-344. 657. Vesikari T, Karvonen A, Korhonen T, et al. Safety and immunogenicity of RIX4414
630. Swartz TA, Handsher R, Stoeckel P, et al. Immunologic memory induced at birth live attenuated human rotavirus vaccine in adults, toddlers and previously
by immunization with inactivated polio vaccine in a reduced schedule. Eur J uninfected infants. Vaccine. 2004;22:2836-2842.
Epidemiol. 1989;5:143-145. 658. Vesikari T, Karvonen A, Prymula R, et al. Efficacy of human rotavirus vaccine
631. Szu SC. Development of Vi conjugate—a new generation of typhoid vaccine. against rotavirus gastroenteritis during the first 2 years of life in European infants:
Expert Rev Vaccines. 2013;12:1273-1286. randomised, double-blind controlled study. Lancet. 2007;370:1757-1763.
632. Tate JE, Mutuc JD, Panozzo CA, et al. Sustained decline in rotavirus detections 659. Vesikari T, Karvonen A, Puustinen L, et al. Efficacy of RIX4414 live attenuated
in the United States following the introduction of rotavirus vaccine in 2006. human rotavirus vaccine in Finnish infants. Pediatr Infect Dis J. 2004;23:937-943.
Pediatr Infect Dis J. 2011;30(suppl):S30-S34. 660. Vesikari T, Matson DO, Dennehy P, et al. Safety and efficacy of a pentavalent
633. Taunay AdE, Galvao PA, deMorais JS, et al. Disease prevention by meningococcal human-bovine (WC3) reassortant rotavirus vaccine. N Engl J Med. 2006;
serogroup C polysaccharide vaccine in preschool children: results after eleven 354:23-33.
months in Sao Paulo, Brazil. Pediatr Res. 1974;8:429A. 661. Vessey S, Chan C, Kuter B, et al. Childhood vaccination against varicella:
634. The National Vaccine Advisory Committee. The measles epidemic. The problems, Persistence of antibody, duration of protection, and vaccine efficacy. J Pediatr.
barriers, and recommendations. The National Vaccine Advisory Committee. 2001;139:297-304.
JAMA. 1991;266:1547-1552. 662. Vestergaard M, Hviid A, Madsen KM, et al. MMR vaccination and febrile
635. Thomas AR, Fiore AE, Corwith HL, et al. Hepatitis B vaccine coverage among seizures: evaluation of susceptible subgroups and long-term prognosis. JAMA.
infants born to women without prenatal screening for hepatitis B virus infection: 2004;292:351-357.
effects of the Joint Statement on Thimerosal in Vaccines. Pediatr Infect Dis J. 663. Villa LL, Costa RL, Petta CA, et al. Prophylactic quadrivalent human papil-
2004;23:313-318. lomavirus (types 6, 11, 16, and 18) L1 virus-like particle vaccine in young women:
636. Thomas RE, Lorenzetti DL, Spragins W, et al. The safety of yellow fever vaccine a randomised double-blind placebo-controlled multicentre phase II efficacy trial.
17D or 17DD in children, pregnant women, HIV+ individuals, and older persons: Lancet Oncol. 2005;6:271-278.
systematic review. Am J Trop Med Hyg. 2012;86:359-372. 664. Vodopija I, Baklaic Z, Hauser P, et al. Reactivity and immunogenicity of
637. Thompson LA, Irigoyen M, Matiz LA, et al. The impact of DTaP-IPV-HB bivalent (AC) and tetravalent (ACW135Y) meningococcal vaccines containing
vaccine on use of health services for young infants. Pediatr Infect Dis J. 2006;25: O-acetyl-negative or O-acetyl-positive group C polysaccharide. Infect Immun.
826-831. 1983;42:599-604.
638. Thompson WW, Shay DK, Weintraub E, et al. Mortality associated with 665. Vu DM, Welsch JA, Zuno-Mitchell P, et al. Antibody persistence 3 years after
influenza and respiratory syncytial virus in the United States. JAMA. 2003;289: immunization of adolescents with quadrivalent meningococcal conjugate vaccine.
179-186. J Infect Dis. 2006;193:821-828.
639. Tischer A, Gerike E. Immune response after primary and re-vaccination 666. Wagner G, Lavanchy D, Darioli R, et al. Simultaneous active and passive
with different combined vaccines against measles, mumps, rubella. Vaccine. immunization against hepatitis A studied in a population of travelers. Vaccine.
2000;18:1382-1392. 1993;11:1027-1032.
640. Toneatto D, Ismaili S, Ypma E, et al. The first use of an investigational multi- 667. Wahdan MH, Rizk F, el Akkad AM, et al. A controlled field trial of a serogroup A
component meningococcal serogroup B vaccine (4CMenB) in humans. Hum meningococcal polysaccharide vaccine. Bull World Health Organ. 1973;48:667-673.
Vaccin. 2011;7:646-653. 668. Walter EB, Neuzil KM, Zhu Y, et al. Influenza vaccine immunogenicity in 6- to
641. Toscani L, Batou M, Bouvier P, et al. Comparison of the efficacy of various 23-month-old children: are identical antigens necessary for priming? Pediatrics.
strains of mumps vaccine: a school survey. Soz Praventivmed. 1996;41:341-347. 2006;118:e570-e578.
642. Tozzi AE, Anemona A, Stefanelli P, et al. Reactogenicity and immunogenicity at 669. Ward B, Boulinanne N, Ratnam S, et al. Cellular immunity in measles vaccine
preschool age of a booster dose of two three-component diphtheria-tetanus- failure: demonstration of measles antigen-specific lymphoproliferative responses
acellular pertussis vaccines in children primed in infancy with acellular vaccines. despite limited serum antibody production after revaccination. J Infect Dis.
Pediatrics. 2001;107:E25. 1995;172:1591-1595.
CHAPTER 245  Active Immunizing Agents 2601.e13

670. Wassilak SG, Oberste MS, Tangermann RH, et al. Progress toward global inter- 694. Witte JJ, Karchmer AW, Case G, et al. Epidemiology of rubella. Am J Dis Child.
ruption of wild poliovirus transmission, 2010-2013, and tackling the challenges 1969;118:107-111.
to complete eradication. J Infect Dis. 2014;210(suppl 1):S5-S15. 695. World Health Organization. Special programme on AIDS and expanded
671. Watson BM, Piercy SA, Plotkin SA, et al. Modified chickenpox in children programme on immunization—joint statement: consultation on human
immunized with the Oka/Merck varicella vaccine. Pediatrics. 1993;91:17-22. immunodeficiency virus (HIV) and routine childhood immunization. Wkly
672. Watson B, Seward J, Yang A, et al. Postexposure effectiveness of varicella vaccine. Epidemiol Rec. 1987;62:297-299.
Pediatrics. 2000;105:84-88. 696. World Health Organization. BCG vaccine. Wkly Epidemiol Rec. 2004;79:33-38.
673. Wattanasri S, Boonthai P, Prasert T. Human rabies after late administration of 697. World Health Organization. WHO position on measles vaccines. Vaccine.
human diploid cell vaccine without hyperimmune serum. Lancet. 1982;2:870. 2009;27:7219-7221.
674. Weibel RE, Neff BJ, Kuter BJ, et al. Live attenuated varicella vaccine: efficacy 698. World Health Organization. Global Advisory Committee on Vaccine Safety, report
trial in healthy children. N Engl J Med. 1984;310:1409-1415. of meeting held 3-4 December 2009. Wkly Epidemiol Rec. 2010;85:29-33.
675. Weibel RE, Stokes J Jr, Buynak EB, et al. Live attenuated mumps-virus vaccine. 3. 699. World Health Organization. Haemophilus influenzae type b (Hib) vaccination
Clinical and serologic aspects in a field evaluation. N Engl J Med. 1967;276:245-251. position paper—July 2013. Wkly Epidemiol Rec. 2013;88:413-426.
676. Weibel RE, Villarejos VM, Klein EB, et al. Clinical and laboratory studies of live 700. World Health Organization. Japanese encephalitis vaccines: WHO position paper,
attenuated RA 27/3 and HPV 77-DE rubella virus vaccines. Proc Soc Exp Biol February 2015. Wkly Epidemiol Rec. 2015;90:69-87.
Med. 1980;165:44-49. 701. World Health Organization. Polio vaccines: WHO position paper, March, 2016.
677. Weinbaum C, Lyerla R, Margolis HS, et al. Prevention and control of infections Wkly Epidemiol Rec. 2016;91:145-168.
with hepatitis viruses in correctional settings. Centers for Disease Control and 702. Wright PF, Cherry JD, Foy HM, et al. Antigenicity and reactogenicity of influenza
Prevention. MMWR Recomm Rep. 2003;52(RR-1):1-36, quiz CE31-34. A/USSR/77 virus vaccine in children—a multicentered evaluation of dosage and
678. Weinberg MS. Vaccine recommendations for infants and children. In: Brunette safety. Rev Infect Dis. 1983;5:758-764.
G, ed. CDC Health Information for International Travel, 2016. New York: Oxford 703. Wright JG, Quinn CP, Shadomy S, et al. Use of anthrax vaccine in the United
University Press; 2016:490-521. States: recommendations of the Advisory Committee on Immunization Practices
679. Weintraub ES, Baggs J, Duffy J, et al. Risk of intussusception after monovalent (ACIP), 2009. MMWR Recomm Rep. 2010;59(RR-6):1-30.
rotavirus vaccination. N Engl J Med. 2014;370:513-519. 704. Wright PF, Thompson J, Vaughn WK, et al. Trials of influenza A/New Jersey/76
680. Wenger JD, Ward JI, Broome CV. Prevention of Haemophilus influenzae type b virus vaccine in normal children: an overview of age-related antigenicity and
disease: vaccines and passive prophylaxis. Curr Clin Top Infect Dis. 1989;10:306-339. reactogenicity. J Infect Dis. 1977;136(suppl):S731-S741.
681. Werzberger A, Kuter B, Nalin D. Six years’ follow-up after hepatitis A vaccination. 705. Xie O, Pollard AJ, Mueller JE, et al. Emergence of serogroup X meningococcal
N Engl J Med. 1998;338:1160. disease in Africa: need for a vaccine. Vaccine. 2013;31:2852-2861.
682. Werzberger A, Mensch B, Kuter B, et al. A controlled trial of a formalin-inactivated 706. Yang YT, Debold V. A longitudinal analysis of the effect of nonmedical exemption
hepatitis A vaccine in healthy children. N Engl J Med. 1992;327:453-457. law and vaccine uptake on vaccine-targeted disease rates. Am J Public Health.
683. Werzberger A, Mensch B, Nalin DR, et al. Effectiveness of hepatitis A vaccine 2014;104:371-377.
in a former frequently affected community: 9 years’ followup after the Monroe 707. Yeh SH, Gurtman A, Hurley DC, et al. Immunogenicity and Safety of
field trial of VAQTA. Vaccine. 2002;20(13-14):1699-1701. 13-Valent Pneumococcal Conjugate Vaccine in Infants and Toddlers. Pediatrics.
684. Wharton M, Cochi SL, Hutcheson RH, et al. A large outbreak of mumps in the 2010;126:e493-e505.
postvaccine era. J Infect Dis. 1988;158:1253-1260. 708. Yih WK, Lieu TA, Kulldorff M, et al. Intussusception risk after rotavirus vaccination
685. White CJ, Kuter BJ, Hildebrand CS, et al. Varicella vaccine (VARIVAX) in healthy in U.S. infants. N Engl J Med. 2014;370:503-512.
children and adolescents: results from 1987-1989 clinical trials. Pediatrics. 709. Yun K, Urban K, Mamo B, et al. Increasing hepatitis B vaccine prevalence among
1991;87:604-610. refugee children arriving in the United States, 2006-2012. Am J Public Health.
686. Whitney CG, Farley MM, Hadler J, et al. Decline in invasive pneumococcal 2016;106:1460-1462.
disease after the introduction of protein-polysaccharide conjugate vaccine. N 710. Yvonnet B, Coursaget P, Deubel V, et al. Simultaneous administration of hepatitis
Engl J Med. 2003;348:1737-1746. B and yellow fever vaccines. J Med Virol. 1986;19:307-311.
687. Giersing BK, Modjarrad K, Kaslow DC, Moorthy VS. Report from the World 711. Zangwill KM, Belshe RB. Safety and efficacy of trivalent inactivated influenza
Health Organization’s Product Development for Vaccines Advisory Committee vaccine in young children: a summary for the new era of routine vaccination.
(PDVAC) meeting, Geneva, September 7-9, 2015. WHO Product Development Pediatr Infect Dis J. 2004;23:189-197.
for Vaccines Advisory Committee; WHO Product Development for Vaccines 712. Zangwill KM, Stout RW, Carlone GM, et al. Duration of antibody response after
Product Development Advisory Committee. Vaccine. 2016;34:2865-2869. meningococcal polysaccharide vaccination in US Air Force personnel. J Infect
688. Widdowson MA, Meltzer MI, Zhang X, et al. Cost-effectiveness and potential Dis. 1994;169:847-852.
impact of rotavirus vaccination in the United States. Pediatrics. 2007;119:684-697. 713. Zell ER, Dietz V, Stevenson J, et al. Low vaccination levels of US preschool and
689. Willis E, Marko A, Marin M, et al. 1048Pregnancy registry for varicella-zoster school-age children. Retrospective assessments of vaccination coverage, 1991-1992.
virus-containing vaccines: 18-year summary of pregnancy outcomes. Open Forum JAMA. 1994;271:833-839.
Infect Dis. 2014;1(suppl 1):S307. 714. Zerboni L, Nader S, Aoki K, et al. Analysis of the persistence of humoral and
690. Winer RL, Lee SK, Hughes JP, et al. Genital human papillomavirus infection: cellular immunity in children and adults immunized with varicella vaccine. J
incidence and risk factors in a cohort of female university students. Am J Epidemiol. Infect Dis. 1998;177:1701-1704.
2003;157:218-226. 715. Zhou F, Harpaz R, Jumaan AO, et al. Impact of varicella vaccination on health
691. Winter K, Glaser C, Watt J, et al. Pertussis epidemic—California, 2014. MMWR care utilization. JAMA. 2005;294:797-802.
Morb Mortal Wkly Rep. 2014;63:1129-1132. 716. Zimmerman RK, Nowalk MP, Chung J, et al. 2014-2015 Influenza vaccine
692. Wise RP, Iskander J, Pratt RD, et al. Postlicensure safety surveillance for 7-valent effectiveness in the United States by vaccine type. Clin Infect Dis. 2016;63:
pneumococcal conjugate vaccine. JAMA. 2004;292:1702-1710. 1564-1573.
693. Wise R, Salive M, Braun M, et al. Postlicensure safety surveillance for varicella 717. Zuckerman AJ. Effect of hepatitis B virus mutants on efficacy of vaccination.
vaccine. JAMA. 2000;284:1271-1279. Lancet. 2000;355:1382-1384.

S-ar putea să vă placă și