You are on page 1of 11

STATE-OF-THE-ART REVIEWS

Booster Vaccinations: Can Immunologic Memory


Outpace Disease Pathogenesis?
AUTHOR: Michael E. Pichichero, MD
Rochester General Research Institute, Rochester General abstract
Hospital, Rochester, New York Almost all current vaccines work by the induction of antibodies in
KEY WORDS serum or on the mucosa to block adherence of pathogens to epithelial
vaccines, immunologic memory, pathogenesis, Neisseria
meningitides, Haemophilus influenzae type b, human papilloma cells or interfere with microbial invasion of the bloodstream. However,
virus antibody levels usually decline after vaccination to undetectable
ABBREVIATIONS amounts if further vaccination does not occur. Persistence of vaccine-
TLR—Toll-like receptor induced antibodies usually goes well beyond the time when they should
Hib—Haemophilus influenzae type b
have decayed to undetectable levels because of ongoing “natural”
DTaP— diphtheria-tetanus-acellular pertussis
HPV— human papilloma virus boosting or other immunologic mechanisms. The production of mem-
www.pediatrics.org/cgi/doi/10.1542/peds.2008-3645 ory B and T cells is of clear importance, but the likelihood that a mem-
doi:10.1542/peds.2008-3645
ory response will be fast enough in the absence of a protective circu-
lating antibody level likely depends on the pace of pathogenesis of a
Accepted for publication Jun 15, 2009
specific organism. This concept is discussed with regard to Haemophi-
Address correspondence to Michael E. Pichichero, MD,
Rochester General Research Institute, Rochester General lus influenzae type b, Streptococcus pneumoniae, and Neisseria men-
Hospital, 1425 Portland Ave, Rochester, NY 14621. E-mail: ingitidis; hepatitis A and B; diphtheria, tetanus, and pertussis; polio,
michael.pichichero@rochestergeneral.org measles, mumps, rubella, and varicella; rotavirus; and human papil-
PEDIATRICS (ISSN Numbers: Print, 0031-4005; Online, 1098-4275). loma virus. With infectious diseases for which the pace of pathogenesis
Copyright © 2009 by the American Academy of Pediatrics is less rapid, some individuals will contract infection before the mem-
FINANCIAL DISCLOSURE: The author has indicated he has no ory response is fully activated and implemented. With infectious dis-
financial relationships relevant to this article to disclose. eases for which the pace of pathogenesis is slow, immune memory
should be sufficient to prevent disease. Pediatrics 2009;124:1633–1641

PEDIATRICS Volume 124, Number 6, December 2009 1633


Downloaded from http://pediatrics.aappublications.org/ by guest on February 24, 2018
With the introduction of so many new TABLE 1 Quantitative Correlates of Protection After Vaccination
vaccines over the past decade and Vaccine Test Correlate of Protection
many more on the horizon, clinicians Hib conjugate ELISA 0.15 ␮g/mL
are often asked, or wonder them- Pneumococcus ELISA; opsonophagocytosis 0.20–0.35 ␮g/mL (for children); 1/8 dilution
Meningococcus Bactericidal 1:8 human complement
selves, whether boosters for all these 1:16 rabbit complement
vaccines will be necessary down the Diphtheria Toxin neutralization 0.01–0.10 IU/mL
road. Sentinel study populations have Tetanus Toxin neutralization 0.01 IU/mL
Acellular pertussis ELISA Unknown
been established by vaccine compa- Polio Microneutralization 1/4–1/8 dilution
nies as phase 4 postlicensure evalua- Measles Microneutralization 120 mIU/mL
tions to forewarn public health author- Mumps ELISA 2 ELU/mL
Rubella Immunoprecipitation 10–15 mIU/mL
ities and practitioners if such boosters Varicella SN; gpELISA ⱖ1/64 dilution; ⱖ5 IU/mL
will be needed. In this review I will Hepatitis B ELISA 10 mIU/mL
present information on what is known Hepatitis A ELISA 10 mIU/mL
Rotavirus ELISA IgA; microneutralization Unknown
about B-cell and T-cell immune mem- HPV ELISA; microneutralization Unknown
ory and innate immunity and the pace ELISA indicates enzyme-linked immunosorbent assay; ELU, ELISA Units; SN, serum neutralization; gpELISA, glyco protein ELISA
of pathogenesis for the diseases we enzyme-linked immunosorbent assay; Ig, immunoglobulin.
Adapted from Plotkin SA. Clin Infect Dis. 2008;47(3):401– 408.
are currently preventing with vaccina-
tion, and I will examine the dynamic
nature of the situation as changes in
many diseases (Table 1). The levels of antibodies should have disappeared
natural boosting occur.
antibody that correlate with protection according to the mathematics of their
IMPORTANCE OF ANTIBODIES are generally derived from population half-life. This may be a result of ongo-
studies wherein observations have ing “natural” boosting or other immu-
As observed by Robbins et al1 and Plot- been made that individuals with a cer- nologic mechanisms discussed below.
kin,2 almost all current vaccines work tain level of antibody are always or
by the induction of antibodies in serum Natural boosting can occur by asymp-
nearly always protected from disease. tomatic colonization by the pathogen
or on the mucosa (by local production However, a specific level of antibody is
or transudation from serum) to block or by a nonpathogen expressing a
not an absolute correlate of protection
adherence of pathogens to epithelial cross-reactive antigen. Natural boost-
for every person, because it is not only
cells or interfere with microbial inva- ing can decrease over time as a patho-
the quantity of antibody that is impor-
sion of the bloodstream. To protect, gen circulates less widely in a popula-
tant but also the functionality of the
the induced antibodies must either be tion because of increasing use of a
antibody. Also, there is genetic varia-
functional against the relevant patho- vaccine and/or the establishment of
tion in susceptibility to disease, differ-
gen or aid the immune system as an herd immunity. This is an ongoing is-
ences in virulence among pathogen
opsonin, or, if the organism exerts its sue relative to several vaccines, be-
strains, differences in innate immune
pathogenic effect by elaborating a responses among individuals, and cause the absence of natural boosting
toxin, then the antibodies must neu- variation in the inoculum of the patho- among vaccine recipients may lead to
tralize that toxin.2 The importance of gen, and there may be an impact a return to disease susceptibility.
antibodies in vaccine-induced protec- from concurrent illness or coinfection. Other explanations have been pro-
tion is undeniable, as supported by Therefore, a specified protective level posed for the persistence of low levels
studies in which passive administra- of antibody should be considered as a of circulating antibodies long after an-
tion prevents or ameliorates disease close estimate applicable in the major- tigen exposure has occurred. One hy-
and the observation of a protective ef- ity of hosts as a relative correlate of pothesis is that small amounts of anti-
fect in the newborn by maternal an- protection. gen are retained and persist inside
tibody.3 All maternal protection of Antibody levels usually decline to unde- peripheral lymph nodes and spleen.
the fetus and newborn occurs as a tectable amounts over time if further The small amount of antigen is enough
result of antibodies, because mater- antigen stimulation does not occur, be- to keep the immune response ongo-
nal B cells and T cells do not cross cause antibodies have a half-life of ing.4 A second hypothesis suggests
the placenta. ⬃30 days. However, persistence of that some memory B cells become se-
Specific levels of antibody have been vaccine-induced antibodies usually questered in the bone marrow sanctu-
correlated with protection against goes well beyond the time when the ary, where they periodically divide, ma-

1634 PICHICHERO
Downloaded from http://pediatrics.aappublications.org/ by guest on February 24, 2018
STATE-OF-THE-ART REVIEWS

ture to plasma cells, and produce whereby a one-fifth to one-tenth stan- A faster and broader memory T-cell re-
small amounts of antibody in serum.5 A dard antigen dose is given to the test sponse has the potential to control in-
third hypothesis is that nonspecific subjects, and then the immune re- fection quickly after reexposure to
polyclonal B-cell activators maintain a sponse is evaluated. This simulation is pathogens.17–19 In both CD4 and CD8 in
continuous, small memory B-cell pool. not perfect, because (1) we do not vitro models the generation of T-cell
The hypothesized nonspecific B-cell know the actual pathogen-specific an- effector functions (cell-mediated im-
activators include several microbial tigen dose, (2) delivery of the antigen by munity) can be evident ⬃2 to 7 days
products that stimulate Toll-like recep- the parenteral route is clearly not the after antigen stimulation.20–23 The
tors (TLRs) (discussed later) and may same as the reality in nature, where maintenance of T-cell memory varies
also occur through bystander T-cell nearly all exposures occur via the skin, according to the antigen. T-cell mem-
help.6,7 respiratory, gastrointestinal, or genito- ory after vaccination with small pro-
urinary tracts, and (3) the context of the tein fragments is much shorter than
ROLE OF MEMORY B CELLS simulation is not during a concurrent ill- that achieved after vaccination with an
ness (eg, a viral upper respiratory infec- attenuated but replicating virus.24–27
The production of memory B cells is a
tion, as often occurs in nature).
complex developmental process that INNATE IMMUNITY
occurs in lymph node and spleen ger- T-CELL MEMORY In addition to adaptive immunity, the
minal centers. During the generation pace of pathogenesis can be slowed by
T-cell immunity plays a role in terminat-
of memory B cells a selection process the innate immune response.28 New in-
ing disease or maintaining protection
occurs called affinity maturation.8 As formation over the past decade has re-
against disease over time. Indeed, T-cell–
antigen becomes less and less avail- vealed that the innate immune system
dependent and antibody-independent
able after vaccination, random muta- has developed a strategy to recognize
mechanisms of protection against infec-
tions of immunoglobulin genes occur conserved structures of microbes that
tions were recently described in mice9–11
and the B cells expressing antibodies are not present on mammalian cells:
and in man.12 Cell immunity is a term
on their surface with the highest affin- so-called pathogen-associated molec-
often used synonymously with T-cell
ity for the diminishing vaccine antigen immunity. ular patterns.29 Also, a recognition and
win out and persist as memory B cells. signaling system called TLRs has been
Two memory T-cell populations are described, which initiates a cascade of
There is a strong correlation between
now known to exist. Effector memory T immunologic events in human host
highly functional antibody and anti-
cells reside in peripheral tissues, cells to contain infection, giving the
body with high affinity for a vaccine an-
whereas another pool, termed central adaptive immune response time to
tigen. The features of vaccine-induced
memory T cells, reside in lymphoid or- respond.30,31 TLR2 is a receptor for
B-cell–mediated immune memory are
gans.13 Both memory T-cell populations lipoteichoic acid of Gram-positive
(1) a rapid production of antibody, (2)
express surface proteins that make bacteria, bacterial lipoproteins, and
predominately immunoglobulin G anti-
them distinguishable from naive T zymosan (yeasts); TLR3 is a receptor
body, (3) a higher antibody level than cells.14 The continuous circulation of
occurred after primary exposure, and for viral double-stranded DNA; TLR4 is
effector memory T cells into tissue is a a receptor for lipopolysaccharides of
(4) production of antibody of higher key feature of the immune system that
affinity (and an antibody population Gram-negative bacteria; and TLR9 is
ensures that memory T cells of partic- a receptor for bacterial DNA.32,33 Once
of higher avidity) for the antigen as a ular specificities are disseminated the TLRs are stimulated by the
result of a process known as affinity throughout the body. pathogen-associated molecular pat-
maturation.
It is well established that memory terns, immunologic effector cells do
To assess the duration of immune CD4⫹ T cells respond more quickly and not need to proliferate or mature; they
memory, vaccinated subjects provide with a wider array of cytokines and can immediately respond by releasing
serum and lymphoid cells for in vitro that memory CD8⫹ T cells respond multiple proinflammatory and antiin-
analysis at time intervals after vacci- with the release of a greater quantity of flammatory mediators and cytokines.
nation. Persistence of antibody lev- cytotoxic molecules (perforin and gran- Because it takes 2 to 7 days for suffi-
els, B memory cells, and T memory zyme B) than occurs during a primary cient amounts of antibodies and effec-
cells can then be measured. The best response.12 Also, memory CD4⫹ T cells tor T cells to be produced, the host re-
tool for assessing immune memory are more effective at helping B cells com- lies on the innate immune system to
is to perform challenge experiments pared with naive CD4 T cells.15,16 hold the infection in check tempo-

PEDIATRICS Volume 124, Number 6, December 2009 1635


Downloaded from http://pediatrics.aappublications.org/ by guest on February 24, 2018
rarily. If the innate response is not ad- for Hib and meningococcus.34–37 In both the United Kingdom after the introduc-
equate, then infection may occur de- cases a detectable response was ob- tion of meningococcal conjugate vac-
spite immune memory. served no sooner than 2 to 7 days after cines. A decrease in antibody despite
antigen exposure. It takes some time persistence of immunologic memory
OUTPACING INFECTION WITH AN was associated with a decline in vac-
from antigen exposure to antibody
IMMUNE RESPONSE cine effectiveness.44–46 As a result of
production, because the antigen must
The likelihood that a B-cell or T-cell be taken up and processed by antigen- the breakthrough cases of disease, the
memory response will be fast enough presenting cells (eg, dendritic cells United Kingdom revised its vaccination
in the absence of a protective circulat- and macrophages); then, the antigen- program to require booster doses of
ing antibody level likely depends in presenting cells must interact with B Hib and meningococcal conjugate vac-
large part on the pace of pathogenesis cells and helper T cells, the B cells must cines so as to maintain protective an-
of the infection caused by a specific proliferate and mature to plasma cells, tibody levels in the blood.
organism (Table 2). Several examples and, finally, the plasma cells release an- After the primary vaccination series in
are described below to illustrate this infancy is completed for Hib conju-
tibody into the circulation.
point.
After introduction of the Hib conjugate gates, only 1 booster is needed in the
Haemophilus influenzae Type b, vaccines, a debate emerged on the second year of life to afford protection
Streptococcus pneumoniae, and protective role of immune memory in until ⬃5 years of age. This is because
Neisseria meningitidis the absence of detectable antibody.38 during the third through the fifth year
This occurred with the introduction of of life children develop “natural” im-
The pace of pathogenesis for encapsu-
diphtheria-tetanus-acellular pertussis munity to Hib induced by colonization
lated respiratory bacteria such as
(DTaP) vaccines combined with Hib of their gastrointestinal tract by a spe-
Haemophilus influenzae type b (Hib),
cies of Escherichia coli that expresses
Streptococcus pneumoniae, and Neis- conjugate vaccines, because several
a polysaccharide capsule (K1 capsule)
seria meningitidis is very rapid. In a DTaP-Hib combination vaccines did not
nearly identical to the Hib polysaccha-
matter of hours bacteria can adhere to produce as high of an antibody level as
ride capsule47 (Fig 1). Natural antibody
the nasopharynx, gain entry to the administration of the 2 separate vac-
also is produced to meningococci and
bloodstream, replicate, and begin to cines.39,40 The debate ended when Hib
pneumococci, but it is currently un-
seed the meninges. Infection by these disease began to occur in England and
known whether the frequency of devel-
encapsulated bacteria is prevented by Wales, where DTaP-Hib combinations
opment of such natural antibody will
the presence of functional antibody di- were licensed and researchers showed
be sufficient to supplement vaccine-
rected to the polysaccharide capsule. that a circulating level of functional an-
induced immunity. The sporadic oc-
The speed of production of measur- tibody was necessary for long-term
currence of meningococcal and
able antibody responses after memory protection for some children.41–43 This pneumococcal disease throughout
B-cell stimulation has been measured same scenario was repeated again in life speaks to the likelihood for the
need for boosters.
TABLE 2 Pace of Pathogenesis, Memory Mechanism, and Need for Boosters
Disease Pace of Live (Attenuated) vs Booster Needed Hepatitis B and A
Pathogenesis Inert
The pace for pathogenesis of hepatitis
Diphtheria Fast Inert Yes
Tetanus Slow Inert Yesa
B and hepatitis A pathogenesis is slow.
Pertussis Fast Inert Yes Clinically, before hepatitis B and hepa-
Polio Slow Inert No titis A vaccines became available, the
Measles Slow Live (attenuated) No
administration of passive antibody in
Mumps Slow Live (attenuated) No
Rubella Slow Live (attenuated) No the form of intramuscular ␥ globulin
Hib Fast Inert Yes could be done 2 weeks after exposure
Hepatitis B Slow Inert No and still be effective. Immunity to hep-
Varicella Slow Live (attenuated) ?
Pneumococcal Fast Inert Yes atitis B and A is lifelong after natural
Meningococcal Fast Inert Yes infection.
Hepatitis A Slow Inert No
Rotavirus Fast Live (reassortant) No
Hepatitis B vaccine–induced antibody
HPV Unknown Inert ? levels wane over time such that by 10
a The booster can be given after exposure, because the pace of pathogenesis is slow. years after vaccination only approxi-

1636 PICHICHERO
Downloaded from http://pediatrics.aappublications.org/ by guest on February 24, 2018
STATE-OF-THE-ART REVIEWS

Diphtheria, Tetanus, and Pertussis


Diphtheria, tetanus, and pertussis are
diseases caused by release of toxins;
there is no bacteremia. The pace of
pathogenesis is several days (diphtheria
and tetanus) to several weeks (pertus-
sis) between infection and the elabora-
tion of sufficient toxin to manifest as dis-
ease. Natural infection does not confer
lifelong protection from any of these 3
diseases. Memory responses to tetanus
have been shown to occur after a time
lag of several days to 2 weeks.50
Breakthrough cases of diphtheria in the
FIGURE 1
Vaccine raises serum antibody levels to Hib, and then serum antibody levels wane. However, “natural” former Soviet Union produced an epi-
serum antibody to the capsular polysaccharide of Hib is produced by colonization of the gastrointes- demic in the 1980s that included many
tinal tract by a strain of E coli that expresses a polysaccharide capsule that is virtually identical to the
Hib capsule, thereby maintaining protection without the need for further boosters. Hib conjugate
vaccinated individuals.51 Breakthrough
indicates Hib polysaccharide conjugated to a protein carrier vaccine. cases of tetanus are well known to oc-
cur among vaccine recipients, thus
leading to the recommended every-10-
year booster.52 Waning immunity has
been demonstrated to occur after use
of whole-cell pertussis vaccines,53–56
leading to adolescent and adult cases
among vaccinated individuals57–59 and
likely will occur in the future after acellu-
lar pertussis vaccines if boosters are not
given. In terms of pathogenesis one
might predict that the prolonged pro-
drome of pertussis, generally 1 to 3
weeks of upper respiratory–like symp-
toms before cough illness begins, should
be sufficient in length to allow immune
memory to outpace disease. However,
this does not occur most likely because
replication of the organism is only on
FIGURE 2
Vaccination raises serum antibody level to hepatitis B surface antigen, and then serum antibody levels the mucosal surface (not in the blood-
wane. After exposure to hepatitis B, a memory antibody response occurs over 2 to 14 days. Because stream) and the organism itself does
the pace of pathogenesis is slow for hepatitis B, the memory response occurs in time to prevent
infection. HbsAg indicates hepatitis B surface antigen; HbsAb, hepatitis B surface antibody.
not elicit a vigorous inflammatory/
immunologic response when it is pres-
ent in the tracheobronchial tree. Per-
haps only with the elaboration of suffi-
mately one third of vaccine recipients report of the persistence of immuno- cient pertussis toxin does the immune
still have detectable antibody levels.48 logic memory 15 to 18 years after hep- system become stimulated.
Nevertheless, up to now there has atitis B vaccination raises con-
been no breakthrough disease. This cerns.49 Less information is available Polio, Measles, Mumps, Rubella,
success has been attributed to mem- about persistence of immunity and and Varicella
ory antibody responses that occur in immune memory after hepatitis A Polio, measles, mumps, rubella, and vari-
sufficient time to afford protection af- vaccination, because it was intro- cella are characterized by 2 viremic
ter exposure (Fig 2). However, a recent duced more recently. phases during pathogenesis. A first vire-

PEDIATRICS Volume 124, Number 6, December 2009 1637


Downloaded from http://pediatrics.aappublications.org/ by guest on February 24, 2018
mia occurs 2 to 4 days after exposure,
and then there is a 1- to 3-day hiatus fol-
lowed by a second and larger viremic
stage during which wider dissemination
of the virus occurs. Thereafter, the onset
of prodrome symptoms occurs, followed
by the classical manifestations of dis-
ease. Permanent immunity is acquired
after natural infection.
For polio, measles, mumps, rubella, and
varicella, the pace of pathogenesis may
be sufficiently slow to allow immune
memory responses to intervene and pre-
vent the important, disease-causing sec-
ond viremic phase in most individuals.
Booster doses of polio, measles, mumps, FIGURE 3
Vaccination raises antibody levels in serum to HPV (that have the potential to transudate onto the
rubella, and varicella vaccines are not mucosa), and then serum antibody levels wane. Persistence of serum antibody above protective levels
currently recommended, but waning im- may occur from natural exposure to HPV (ie, natural boosting or other immunologic mechanisms).
munity has been raised as a concern.60–64
Additional doses of these vaccines have childhood, because the infection rate best surrogate marker for protection
been suggested to produce immunity is much lower in older children. from HPV infection after vaccination.84
among a relatively small cohort of indi-
viduals who fail to respond to primary Human Papilloma Virus CONCLUSIONS
vaccination.65,66 Live, attenuated strains The mechanism of protection after hu-
as opposed to killed viral vaccines more It generally requires ⬃2 to 5 days for B
man papilloma virus (HPV) vaccination
closely mimic natural infection where memory cells and T memory cells (cell-
seems to be the production of neutraliz-
immunity is known to be lifelong. It is mediated immunity) to expand and give
ing antibody.69–72 The pace of pathogene-
possible that reactivation of latent vac- rise to mature immune effector cells af-
sis for HPV in humans is not known. In
cine virus induces boosts in antibody ter a host experiences exposure to a po-
animal models it takes 30 minutes to
levels. The duration of immunity from en- tential pathogen. The innate immune sys-
24 hours73–77 for the virus to gain entry
hanced inactivated polio vaccine is un- tem and preexisting circulating antibody
to the basal epithelial cells (and infect
der prospective study.67 levels must prevent progression of dis-
the cell); thereafter, the virus becomes
largely inaccessible to antibody. There- ease until memory responses occur.
Rotavirus fore, it would seem that a minimum level With infectious diseases for which the
Natural rotavirus infections are not of antibody would need to be present in pace of pathogenesis is rapid, some indi-
fully protective, although reinfections mucus (whether locally produced or as a viduals will contract infection before the
are uniformly milder in severity than result of transudation from serum) at memory response is fully activated and
primary infections.68 The 2 currently the time of exposure to prevent infection implemented. With infectious diseases
available rotavirus vaccines are live at- (Fig 3). After HPV vaccination, it is cur- for which the pace of pathogenesis is
tenuated reassortant vaccines. The rently uncertain whether “natural” cer- slow, immune memory should be suffi-
pace of pathogenesis of rotavirus in- vical HPV infection would be sufficient cient to prevent disease. For some newer
volves several days from infection to to stimulate a protective antibody re- vaccines there is uncertainty whether
disease. Although immunity may be in- sponse (natural boosting).78–83 Although breakthrough infections will occur. This
complete after infection and may wane the role of immune memory remains un- is an area of active research that will
after vaccination, it is unlikely that certain, high and sustained neutralizing provide answers to this important ques-
boosters will be recommended in later antibody titers are considered to be the tion in the future.

1638 PICHICHERO
Downloaded from http://pediatrics.aappublications.org/ by guest on February 24, 2018
STATE-OF-THE-ART REVIEWS

REFERENCES
1. Robbins JB, Schneerson R, Szu S. Perspective: for vaccine development. Nat Rev Immunol. Ahmed R. Cytotoxic T cell memory without
hypothesis—serum IgG antibody is sufficient 2002;2(4):251–262 antigens. Nature. 1994;369(6482):648 – 652
to confer protection against infectious dis- 14. Okada R, Kondo T, Matsuki F, Takata H, 27. Swain SL, Croft MC, Dubey C, et al. From na-
eases by inactivating the inoculum. J Infect Takiguchi M. Phenotypic classification of ive to memory T cells. Immunol Rev. 1996;
Dis. 1995;171(6):1387–1398 human CD4⫹ T cell subsets and their differ- 150:143–167
2. Plotkin SA. Vaccines: correlates of vaccine- entiation. Int Immunol. 2008;20(9): 28. Akira S, Uematsu S, Takeuchi O. Pathogen
induced immunity. Clin Infect Dis. 2008; 1189 –1199 recognition and innate immunity. Cell. 2006;
47(3):401– 408 15. Swain SL. Generation and in vivo persis- 124(4):783– 801
3. Zinkernagle RM, Hengartner H. Protective tence of polarized Th1 and Th2 memory 29. Medzhitov R, Janeway C Jr. Innate immunity.
“immunity” by pre-existent neutralizing an- cells. Immunity. 1994;1(7):543–552 N Engl J Med. 2000;343(5):338 –344
tibody titers and preactivated T cells but not 16. Croft M, Swain SL. Analysis of CD4⫹ T cells 30. Ulevitch RJ. Regulation of receptor-
by so-called “immunological memory.” Im- that provide contact-dependent bystander dependent activation of the innate immune
munol Rev. 2006;211:310 –319 help to B Cells. J Immunol. 1992;149(10): response. J Infect Dis. 2003;187(suppl 2):
4. Bachmann MT, Odermat B, Hengertner H, 3157–3165 S351–S355
Zinkernagel RM. Induction of long-lived ger- 17. Bachmann MF, Barner M, Viola, Kopf M. Dis- 31. Pulendran B. Modulating vaccine responses
minal centers associated with persisting tinct kinetics of cytokine production and cy- with dendritic cells and Toll-like receptors.
antigen after viral infection. J Exp Med. tolysis in effector and memory T cells after Immunol Rev. 2004;199:227–250
1996;183(5):2259 –2269 viral infection. Eur J Immunol. 1999;29(1): 32. Beutler B. Inferences, questions and possi-
5. Osmond DG. Population dynamics of bone 291–299 bilities in Toll-like receptor signalling. Na-
marrow B lymphocytes. Immunol Rev. 1986; 18. Viega-Fernandez H, Walter U, Bourgeois C, ture. 2004;430(6996):257–263
93:103–124 McLean A, Rocha B. Response of naïve and 33. Netea MG, Vand der Meer JWN, Sutmuller
6. Bernasconi N, Traggiai E, Lanzavecchia A. memory CD8⫹ T cells to antigen stimulation RP, Adema GJ, Kullberf B. From the Th1/Th2
Maintenance of serological memory by in vivo. Nat Immunol. 2000;1(1):47–53 paradigm towards a Toll-like receptor/T-
polyclonal activation of human memory B 19. Kaech SM, Ahmed R. Memory CD8⫹ cells helper bias. Antimicrob Agents Chemother.
cells. Science. 2002;298(5601):2199 –2202 differentiation: initial antigen encounter 2005;49(10):3991–3996
7. Traggiai E, Puzone R, Lanzavecchia A. Anti- triggers a developmental program in naïve 34. Pichichero ME, Voloshin T, Passador S. Ki-
gen dependent and independent mecha- cells. Nat Immunol. 2001;2(5):415– 422 netics of booster responses to Haemophi-
nisms that sustain serum antibody levels. 20. Bradley LM, Duncan DD, Tonkonogy SL, lus influenzae type b conjugate after
Vaccine. 2003;21(suppl 2):S35–S37 Swain SL. Characterization of antigen- combined diphtheria-tetanus-acellular per-
specific CD4⫹ effector T cells in vivo: immu- tussis Haemophilus influenzae type b vacci-
8. Andersson K, Wrammert J, Leanderson T.
nization results in a transient population of nation in infants. Pediatr Infect Dis J. 1999;
Affinity selection and repertoire shift: para-
MEL⫺14⫺, CD45RB-helper cells that se- 18(12):1106 –1108
doxes as a consequence of somatic muta-
tion? Immunol Rev. 1998;162:173–182 cretes interleukin 2 (IL-2), IL-3, IL-4, and in- 35. Madore DV, Johnson-Kraines CL, Rothstein
terferon gamma. J Exp Med. 1991;174(3): EP, Smith DH. Kinetics of antibody response
9. McCool TL, Weiser JN. Limited role of anti-
547–559 to Haemophilus influenzae type B vaccines.
body in clearance of Streptococcus pneu-
Curr Med Res Opin. 1999;15(2):105–112
moniae in a murine model of colonization. 21. Swain SL, Weinberg AD, English M, Huston G.
Infect Immun. 2004;72(10):5807–5813 IL-4 directs the development of Th2-like 36. Snape MD, Kelly DF, Salt P, et al. Serogroup C
helper effectors. J Immunol. 1990;145(11): meningococcal glycoconjugate vaccine in
10. Malley R, Trzcinski K, Srivastava A, Thomp-
3796 –3806 adolescents: persistence of bactericidal an-
son CM, Anderson PW, Lipsitch M. CD4⫹ T tibodies and kinetics of the immune re-
cells mediate antibody-independent ac- 22. Azuma M, Cayabyab M, Phillips JH, Lanier LL.
sponse to a booster vaccine more than 3
quired immunity to pneumococcal coloniza- Requirements for CD28-dependent T cell-
years after immunization. Clin Infect Dis.
tion. Proc Natl Acad Sci USA. 2005;102(13): mediated cytotoxicity. J Immunol. 1993;
2006;43(11):1387–1394
4848 – 4853 150(6):2091–2101
37. Tsai TF, Borrow R, Gnehm E, et al. Early ap-
11. Trzcinski K, Thompson CM, Srivastava A, 23. Bradley LM, Duncan DD, Yoshimoto K, Swain
pearance of bactericidal antibodies after
Basset A, Malley R, Lipsitch M. Protection SL. Memory effectors: a potent, IL-4 secret-
polysaccharide challenge of toddlers
against nasopharyngeal colonization by ing helper T cell population that develops primed with a group C meningococcal con-
Streptococcus pneumoniae is mediated by after restimulation with antigen. J Immu- jugate vaccine: what is its role in the main-
antigen-specific CD4⫹ T cells. Infect Immun. nol. 1993;150(8 pt 1):3119 –3130 tenance of protection? Clin Vaccine Immu-
2008;76(6):2678 –2684 24. Gray D, Matzinger P. T cell memory is short- nol. 2006;13(8):854 – 861
12. Zhang Q, Bagrade L, Bernatoniene J, et al. lived in the absence of antigen. J Exp Med. 38. Anderson P, Ingram DL, Pichichero ME, Pe-
Low CD4 T cell immunity to pneumolysin is 1991;174(5):969 –974 ter G. A high degree of natural immunologic
associated with nasopharyngeal carriage 25. Oehen S, Waldner H, Kundig TM, Hengartner priming to the capsular polysaccharide
of pneumococci in children [published cor- H, Zinkernagel RM. Antivirally protective cy- may not prevent Haemophilus influenzae
rection appears in J Infect Dis. 2008;197(9): totoxic T cell memory to lymphocytic chorio- type b meningitis. Pediatr Infect Dis J. 2000;
1354]. J Infect Dis. 2007;195(8):1194 –1202 meningitis virus is governed by persisting 19(7):589 –591
13. Kaech SM, Wherry EJ, Ahmed R. Effector and antigen. J Exp Med. 1992;176(5):1273–1281 39. Eskola J, Olander R, Hovi T, Litmanen L, Pel-
memory T-cell differentiation: implications 26. Lau LL, Jamieson BD, Somasundaram T, tola S, Kayhty H. Randomised trial of the ef-

PEDIATRICS Volume 124, Number 6, December 2009 1639


Downloaded from http://pediatrics.aappublications.org/ by guest on February 24, 2018
fect of co-administration with acellular per- tion of humans. Cell Immunol. 1979;45(1): strain of live varicella vaccine. Pediatrics.
tussis DTP vaccine on immunogenicity of 142–150 1985;75(4):667– 671
Haemophilus influenzae type b conjugate 51. Vitek CR, Brennan MB, Gotway CA, et al. Risk 64. Anderson LJ, Seward JF. Mumps epidemiol-
vaccine. Lancet. 1996;348(9043):1688 –1692 of diphtheria among schoolchildren in the ogy and immunity: the anatomy of a modern
40. Rennels MB, Englund J, Bernstein D, et al. Dim- Russian Federation in relation to time since epidemic. Pediatr Infect Dis J. 2008;27(10
inution of the anti-polyribosylribitol phos- last vaccination. Lancet. 1999;353(9150): suppl):S75–S79
phate response to a combined diphtheria- 355–358 65. Lopez AS, Guris D, Zimmerman L, et al. One
tetanus-acellular pertussis/Haemophilus 52. Gergen P, McQuillan GM, Kiely M, Ezzati-Rice dose of varicella vaccine does not prevent
influenzae type b vaccine by concurrent inac- TM, Sutter RW, Virella G. A population-based school outbreaks: is it time for a second
tivated poliovirus vaccination. Pediatr Infect serologic survey of immunity to tetanus in dose? Pediatrics. 2006;117(6). Available at:
Dis J. 2000;19(5):417– 422 the United States. N Engl J Med. 1995; www.pediatrics.org/cgi/content/full/117/
41. McVernon J, Johnson PD, Pollard AJ, Slack 332(12):761–766 6/e1070
MP, Moxon ER. Immunologic memory in Hae- 53. Heininger U. Recent progress in clinical and 66. Arvin A, Gershon A. Control of varicella: why
mophilus influenzae type b conjugate vac- basic pertussis research. Eur J Pediatr. is a two-dose schedule necessary? Pediatr
cine failure. Arch Dis Child. 2003;88(5): 2001;160(4):203–213 Infect Dis J. 2006;25:475– 476
379 –383 67. Vidor E, Meschievitz C, Plotkin S. Fifteen years
54. Jansen DL, Gray GC, Putnam SD, Lynn F,
42. Oh SY, Griffiths D, John T, et al. School-aged Meade BD. Evaluation of pertussis in U.S. of experience with Vero-produced enhanced
children: a reservoir for continued circula- Marine Corps trainees. Clin Infect Dis. 1997; potency inactivated poliovirus vaccine. Pedi-
tion of Haemophilus influenzae type b in the 25(5):1099 –1107 atr Infect Dis J. 1997;16(3):312–322
United Kingdom. J Infect Dis. 2008;197(9): 68. Glass RI, Parashar UD, Bresee JS, et al. Rotavi-
55. Cattaneo LA, Reed GW, Haase DH, Wills MJ,
1275–1281 rus vaccines: current prospects and future
Edwards KM. The seroepidemiology of Bor-
43. Yeh CL, Kelly D, Ly-Mee Y, et al. Haemophilus challenges. Lancet. 2006;368(9532):323–332
detella pertussis infections: a study of per-
influenzae type b vaccine failure in children 69. Pastrana DV, Vass WC, Lowy DR, Schiller JT.
sons ages 1– 65 years. J Infect Dis. 1996;
is associated with inadequate production of NHPV16 VLP vaccine induces human anti-
173(5):1256 –1259
high-quality antibody. Clin Infect Dis. 2008; bodies that neutralize divergent variants of
46(2):186 –192 56. Esposito S, Agliardi T, Giammanco A, et al.
HPV16. Virology. 2001;279(1):361–369
Long-term pertussis-specific immunity af-
44. Trotter CL, Andrews NJ, Kaczmarski B, Miller 70. Roden RB, Kirnbauer, Booy FP, Jessie J,
ter primary vaccination with a combined
E, Ramsay ME. Effectiveness of meningococ- Lowy DR, Schiller JT. In vitro generation and
diphtheria, tetanus, tricomponent acellular
cal serogroup C conjugate vaccine 4 years type-specific neutralization of a human pap-
pertussis, and hepatitis B vaccine in com-
after introduction. Lancet. 2004;364(9431): illomaviruses with the cell surface. J Virol.
parison with that after natural infection. In-
365–367 1996;70(9):5875–5883
fect Immun. 2001;69(7):4516 – 4520
45. MacLennan JM, Shackley F, Heath PT, et al. 71. Wang Z, Christensen N, Schiller JT, Dillner J.
57. Mancuso JD, Snyder A, Stigers J, et al. Per-
Safety, immunogenicity, and induction of A monoclonal antibody against intact hu-
tussis outbreak in a US military community:
immunologic memory by a serogroup C me- man papillomavirus type 16 capsids blocks
Kaiserslauten, Germany, April–June 2005.
ningococcal conjugate vaccine in infants: a the serological reactivity of most human
Clin Infect Dis. 2007;45(11):1476 –1478
randomized controlled trial. JAMA. 2000; sera. J Gen Virol. 1997;78(pt 9):2209 –2215
283(21):2795–2801 58. Gilberg S, Njamkepo E, Parent du Chatelet I,
72. Day PM, Gambhira R, Roden RBS, Lowy DR,
et al. Evidence of Bordetella pertussis infec-
46. Snape M, Kelly DF, Green B, Moxon ER, Borrow Schiler JT. Mechanisms of human papillo-
tion in adults presenting with persistent
R, Pollard AJ. Lack of serum bactericidal activ- mavirus type 16 neutralization by l2 cross-
cough in a French area with very high
ity in preschool children two years after a sin- neutralizing and l1 type-specific antibodies.
gle dose of serogroup C meningococcal whole-cell vaccine coverage. J Infect Dis. J Virol. 2008;82(9):4638 – 4646
polysaccharide-protein conjugate vaccine. Pe- 2002;186(3):415– 418
73. Zhou J, Gissman L, Zentgraf H, Muller H,
diatr Infect Dis J. 2005;24(2):128 –131 59. Schmitt-Grohé S, Cherry JD, Heininger U, et Picken M, Muller M. Early phase in the infec-
47. Schneerson R, Robbins J. Induction of se- al. Pertussis in German adults. Clin Infect tion of cultured cells with papillomavirus
rum Haemophilus influenzae type b capsu- Dis. 1995;21(4):860 – 866 virions. Virology. 1995;214(1):167–176
lar antibodies in adult volunteers fed cross 60. Bin C, Zhihui C, Qichang L, et al. Duration of 74. Liu WJ, Wi YM, Shao, KN, Liu YH, Liu XS, Frazer
reacting Escherichia coli. N Engl J Med. immunity following immunization with live IH. Association of bovine papillomavirus
1975;292(21):1093–1096 measles vaccine: 15 years of observation in type 1 with microtubules. Virology. 2001;
48. Lin YC, Chang MH, Ni YH, Hsu HY, Chen DS. Zhejiang Province, China. Bull World Health 282(2):237–244
Long-term immunogenicity and efficacy of Organ. 1991;69(4):415– 423 75. Sibbet G, Romero-Graillet C, Meneguzzi G,
universal hepatitis b virus vaccination in 61. Stittelaar K, De Swart RL, Osterhaus A. Vac- Campo MS. ␣ 6 integrin is not the obligatory
Taiwan. J Infect Dis. 2003;187(1):134 –138 cination against measles: a neverending cell receptor for bovine papillomavirus type
49. Lu CY, Ni YH, Chiang BL, et al. Humoral and story. Expert Rev Vaccines. 2002;1(2): 4 [published correction appears in J Gen
cellular immune responses to a hepatitis B 151–159 Virol. 2000;81(pt 6):1629]. J Gen Virol. 2000;
vaccine booster 15–18 years after neonatal 62. Cohen C, White JM, Savage EJ, et al. Vaccine 81(pt 2):327–334
immunization. J Infect Dis. 2008;197(10): effectiveness estimates, 2004 –2005 76. Ozbun MA. Infectious human papillomavirus
1419 –1425 mumps outbreak, England. Emerg Infect type 31b: purification and infection of an im-
50. Stevens RH, Saxon A. Differential synthesis Dis. 2007;13(1):12–17 mortalized human keratinocyte cell line.
of IgM and IgG anti-tetanus toxoid antibody 63. Asano Y, Nagai T, Miyata T, et al. Long-term J Gen Virol. 2002;83(pt 11):2753–2763
in vitro following in vivo booster immuniza- protective immunity of recipients of the OKA 77. Culp TD, Christensen ND. Kinetics of in vitro

1640 PICHICHERO
Downloaded from http://pediatrics.aappublications.org/ by guest on February 24, 2018
STATE-OF-THE-ART REVIEWS

adsorption and entry of papillomavirus viri- Natural history of human papillomavirus papillomavirus types 16 and 18 in young
ons. Virology. 2004;319(1):152–161 type 16 virus-like particle antibodies in women: a randomised controlled trial. Lan-
78. Stanley M. Immune responses to human young women. Cancer Epidemiol Biomark- cet. 2004;364(9447):1757–1765
papillomavirus. Vaccine. 2006;24:(suppl 1): ers Prev. 2004;13(1):110 –116 83. Villa LL, Costa RL, Petta CA, et al. High sustained
S16 –S22 81. Prakash S, Patterson S, Kapembwa MS. efficacy of a prophylactic quadrivalent human
79. Carter JJ, Koutsky LA, Hughers JP, et al. Macrophages are increased in cervical ep- papillomavirus types 6/11/16/18 L1 virus-like
Comparison of human papillomavirus types ithelium of women with cervicitis. Sex particle vaccine through 5 years of follow-up.
16, 18, and 6 capsid antibody responses fol- Transm Infect. 2001;77(5):366 –369 Br J Cancer. 2006;95(11):1459 –1466
lowing incident infection. J Infect Dis. 2000; 82. Harper DM, Franco EL, Wheeler C, et al. Effi- 84. Stanley M, Lowy DR, Frazer I. Prophylactic
181(6):1911–1919 cacy of a bivalent L1 virus-like particle vac- HPV vaccines: underlying mechanisms. Vac-
80. Ho GYF, Studentsov YY, Bierman R, Burk RD. cine in prevention of infection with human cine. 2006;24(suppl 3):S106 –S113

Rating, Ratings: The increasing public availability of rankings and ratings of physicians and
hospitals is gaining attention in the social media. According to a recent article (Maney K. The
Atlantic. July/August, 2009, p. 38), 47% of internet users now search online for information
about doctors, which can include ratings of such measures as waiting times or medical/
surgical errors. As a result, about 2000 physicians have thus far sought assistance from a
company called Medical Justice that provides advice on the legal rights of physicians who
want to stop some of the ratings being made available to the public. Despite these concerns,
public information on physician ratings—if it is valid and reliable—may be just what the
doctor ordered when it comes to improving the overall quality of our medical care.
Noted by JFL, MD

PEDIATRICS Volume 124, Number 6, December 2009 1641


Downloaded from http://pediatrics.aappublications.org/ by guest on February 24, 2018
Booster Vaccinations: Can Immunologic Memory Outpace Disease Pathogenesis?
Michael E. Pichichero
Pediatrics 2009;124;1633
DOI: 10.1542/peds.2008-3645 originally published online November 23, 2009;

Updated Information & including high resolution figures, can be found at:
Services http://pediatrics.aappublications.org/content/124/6/1633
References This article cites 83 articles, 21 of which you can access for free at:
http://pediatrics.aappublications.org/content/124/6/1633.full#ref-list-
1
Subspecialty Collections This article, along with others on similar topics, appears in the
following collection(s):
Infectious Disease
http://classic.pediatrics.aappublications.org/cgi/collection/infectious_
diseases_sub
Vaccine/Immunization
http://classic.pediatrics.aappublications.org/cgi/collection/vaccine:im
munization_sub
Permissions & Licensing Information about reproducing this article in parts (figures, tables) or
in its entirety can be found online at:
https://shop.aap.org/licensing-permissions/
Reprints Information about ordering reprints can be found online:
http://classic.pediatrics.aappublications.org/content/reprints

Pediatrics is the official journal of the American Academy of Pediatrics. A monthly publication, it
has been published continuously since . Pediatrics is owned, published, and trademarked by the
American Academy of Pediatrics, 141 Northwest Point Boulevard, Elk Grove Village, Illinois,
60007. Copyright © 2009 by the American Academy of Pediatrics. All rights reserved. Print ISSN:
.

Downloaded from http://pediatrics.aappublications.org/ by guest on February 24, 2018


Booster Vaccinations: Can Immunologic Memory Outpace Disease Pathogenesis?
Michael E. Pichichero
Pediatrics 2009;124;1633
DOI: 10.1542/peds.2008-3645 originally published online November 23, 2009;

The online version of this article, along with updated information and services, is
located on the World Wide Web at:
http://pediatrics.aappublications.org/content/124/6/1633

Pediatrics is the official journal of the American Academy of Pediatrics. A monthly publication, it
has been published continuously since . Pediatrics is owned, published, and trademarked by the
American Academy of Pediatrics, 141 Northwest Point Boulevard, Elk Grove Village, Illinois,
60007. Copyright © 2009 by the American Academy of Pediatrics. All rights reserved. Print ISSN:
.

Downloaded from http://pediatrics.aappublications.org/ by guest on February 24, 2018

You might also like