Download as pdf or txt
Download as pdf or txt
You are on page 1of 21

Expert Review of Vaccines

ISSN: (Print) (Online) Journal homepage: www.tandfonline.com/journals/ierv20

The durability of vaccine-induced protection: an


overview

Vipin M. Vashishtha & Puneet Kumar

To cite this article: Vipin M. Vashishtha & Puneet Kumar (2024) The durability of
vaccine-induced protection: an overview, Expert Review of Vaccines, 23:1, 389-408, DOI:
10.1080/14760584.2024.2331065

To link to this article: https://doi.org/10.1080/14760584.2024.2331065

© 2024 The Author(s). Published by Informa


UK Limited, trading as Taylor & Francis
Group.

Published online: 20 Mar 2024.

Submit your article to this journal

Article views: 2718

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ierv20
EXPERT REVIEW OF VACCINES
2024, VOL. 23, NO. 1, 389–408
https://doi.org/10.1080/14760584.2024.2331065

REVIEW

The durability of vaccine-induced protection: an overview


a
Vipin M. Vashishtha and Puneet Kumarb
a
Department of Pediatrics, Mangla Hospital & Research Center, Shakti Chowk, Bijnor, Uttar Pradesh, India; bDepartment of Pediatrician, Kumar
Child Clinic, New Delhi, India

ABSTRACT ARTICLE HISTORY


Introduction: Current vaccines vary widely in both their efficacy against infection and disease, and the Received 18 January 2024
durability of the efficacy. Some vaccines provide practically lifelong protection with a single dose, while Accepted 12 March 2024
others provide only limited protection following annual boosters. What variables make vaccine-induced
KEYWORDS
immune responses last? Can breakthroughs in these factors and technologies help us produce vaccines
Durability; immunity;
with better protection and fewer doses? The durability of vaccine-induced protection is now a hot area immune responses;
in vaccinology research, especially after COVID-19 vaccines lost their luster. It has fueled discussion on persistence of immunity;
the eventual utility of existing vaccines to society and bolstered the anti-vaxxer camp. To sustain public vaccines; vaccinology
trust in vaccines, lasting vaccines must be developed.
Areas covered: This review summarizes licensed vaccines’ protection. It analyses immunological
principles and vaccine and vaccinee parameters that determine longevity of antibodies. The review
concludes with challenges and the way forward to improve vaccine durability.
Expert opinion: Despite enormous advances, we still lack essential markers and reliable correlates of
lasting protection. Most research has focused on humoral immune responses, but we must also focus
on innate, mucosal, and cellular responses – their assessment, correlates, determinants, and novel
adjuvants. Suitable vaccine designs and platforms for durable immunity must be found.

1. Introduction 2. Overview of the durability of protection


conferred by the key licensed vaccines
‘Waning of immunity’ became a buzzword not only in academia
and industry but also among the lay public when it became clear To perform an analysis of the durability of immune protection
that the protection conferred by COVID-19 vaccines is not as accorded by the currently licensed vaccines, we have broadly
durable as offered by other vaccines like measles. The excitement classified them under three heads (Table 1):
over the speedy development and rapid deployment of COVID-19
vaccines gradually waned off, and soon, a debate surrounding the (1) Vaccines that provide long duration of protection (>20
durability of immune protection ensued. Thus, the durability of years).
vaccine-induced protection is a hot area of research today. What (2) Vaccines that provide moderate duration of protection
factors determine the durability of vaccine-induced immune pro­ (5-20 years).
tection? Does the nature of the pathogen (fast mutating virus vs. (3) Vaccines that provide short duration of protection (<5
‘stable’ virus), the biology of the vaccine (live attenuated vs. inacti­ years).
vated viral vaccine, the addition of an adjuvant and the type), or
the vaccination schedule (dose, number of doses and interval
between doses) have any bearing on the long term persistence
of vaccine-derived immunity? It is also known that natural infec­ 2.1. Vaccines that provide a long duration of protection
tions caused by most human coronaviruses, including Severe (>20 years)
Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV2) and influ­ 2.1.1. Measles vaccines
enza, fail to provide long-lasting protection, unlike other respira­ A single dose of vaccine offers lifelong protection to nearly all
tory pathogens like measles, rubella, varicella, and mumps [1]. The vaccinees. The primary immune response is boosted with
current generation of vaccines against all these pathogens also exposure to wild viruses or after re-vaccination [2]. Once ser­
follows a similar pattern.
oconverted, protective anti-measles antibodies ≥ 120 mIU/mL
This review provides an overview of the durability of protection
are detected for a very long period, probably for life, without
offered by the currently used licensed vaccines. It summarizes the
current knowledge, recent advances, knowledge gaps, challenges, any significant waning in the antibody titers. A modeling
and bottlenecks regarding the long-term protection induced by study estimated the half-life of anti-measles antibodies to be
vaccines and how to overcome these. more than 3000 years [3].

CONTACT Vipin M. Vashishtha vipinipsita@gmail.com Director and Consultant Pediatrician, Department of Pediatrics, Mangla Hospital & Research Center,
Shakti Chowk, Bijnor, Uttar Pradesh 246701, India
© 2024 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (http://creativecommons.org/licenses/by-nc/4.0/), which permits
unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited. The terms on which this article has been published allow the
posting of the Accepted Manuscript in a repository by the author(s) or with their consent.
390 V. M. VASHISHTHA AND P. KUMAR

30 years by a single or two-dose vaccine schedule [12,13].


Article highlights Furthermore, protection against the clinical disease may per­
● Only a handful of currently licensed vaccines provide long-lasting
sist even without detectable antibodies in the presence of
protection memory B and T cells [14].
● There are still gaps in our understanding of reliable correlates of
durable protection.
● Induction of vigorous germinal center reaction and induction of
a large number of long-lasting plasma cells along with the formation 2. Vaccines that provide moderate duration of
of memory B-cells and memory T-cells is essential for durable vac­ protection (5–20 years)
cine-induced protection
● There are several determinants of the longevity of antibody response, 2.2.1. Oral polio vaccine (OPV)
such as vaccine platform, target pathogen, vaccinee’s characteristics, After seroconversion, OPV offers almost lifelong protection
environmental and genetic factors
● New developments in the field of vaccinology, like improved vaccine against paralytic poliomyelitis, even if the antibody titers fall
design, novel adjuvants, and molecular delineation of antigenic below detectable levels later in life. However, seroconversion
forms, hold hope for developing durable vaccines and efficacy vary between different geographical settings:
● Further research is needed to identify the optimal antigen delivery
methods and formulations that result in prolonged antigen availabil­ higher in temperate climates in industrialized countries than
ity and can elicit long-lasting and protective immunity in developing countries with tropical conditions. According to
an Italian study, immune protection against paralytic disease
persisted up to 18 years after administration of the last OPV
dose, with protective antibodies against poliovirus type 1 and
2.1.5. Rubella vaccine 2 detected in more than 95% of the subjects even after 30
Rubella vaccine also induces a robust immune response that is years of the last OPV dose [15].
presumed to be lifelong [4]. Rubella virus-specific humoral
immune responses wane with time after vaccination, but they 2.2.2. Inactivated poliovirus vaccine (IPV)
do so moderately [5]. Though no absolute immune correlate of Studies from industrialized countries have indicated that IPV-
protection has been established, a cutoff level of anti-rubella induced antibodies protect from paralytic polio for many dec­
IgG antibodies of ≥ 10 IU/mL is generally protective against ades, probably for life [16]. According to a retrospective cohort
rubella disease [4]. One longitudinal study estimated that study from Italy, neutralizing antibodies against all three polio
rubella-specific immunity was maintained, with an estimated types persisted in more than 90% of study participants 18
half-life of 114 years and no significant rate of titer decrease [3]. years after the last vaccine dose [17]. In this study, the IPV
induced higher titers of neutralizing antibodies than OPV.
2.1.3. Yellow fever vaccine
The live-attenuated yellow fever vaccine has excellent long- 2.2.3. Hepatitis A live attenuated vaccine
term effectiveness. The titer of neutralizing antibodies is the Like inactivated vaccines, live attenuated hepatitis-A vaccines
correlate of protection. Plaque reduction neutralization test also offer a long duration of protection. After a single dose of
(PRNT50) assays with a titer of 1 in 10 are used in most clinical H2 strain-based live attenuated vaccine in an endemic region,
trials to correlate protection. at least 15–17 years of protection has been demonstrated
According to Poland et al. [6], 80.6% of World War II along with the persistence of immune memory [18–20].
soldiers had neutralizing antibodies that lasted longer than
thirty years. Neutralizing antibodies have also shown long- 2.2.4. Human papilloma virus (HPV) vaccines
term persistence in several additional studies conducted in These vaccines are unique in that they offer long-lasting pro­
both endemic and non-endemic countries [7–10]. Since tection against HPV-related illnesses even with a single dose,
there are no documented cases of Yellow Fever infection and the protection they offer is virtually sterilizing, meaning
in individuals with documented seroconversion, it has been that they prevent infection rather than just illness [21]. This is
argued that the hypothesis that the vaccine provides life­ despite them being non-live attenuated vaccines. Bivalent,
long protection is correct [11]. quadrivalent, and nonavalent vaccines have shown the most
extended durations of preventative effect: 11 years in the
Costa Rica trial [22], 14 years in the FUTURE II trial [23], and
2.1.4. Hepatitis-B vaccine
eight years in the long-term follow-up (LTFU) extension study
The Hepatitis B vaccine is a T-cell-dependent subunit vaccine, [24] and Scandinavian Study [25]. An Australian real-world
and hepatitis B infection has a long incubation period, allow­ effectiveness study [26] has the longest observation time of
ing enough time for an anamnestic response to occur if there 12 years for vaccine efficacy (for the quadrivalent vaccine).
is contact with the virus. Hence, once seroconversion occurs,
the vaccine can provide long-term protection even if antibody 2.2.5. Bacille Calmette-Guérin (BCG) vaccine
titers fall below the protective level of 10mIU/mL. Even while the BCG vaccine’s efficacy varies greatly depending
on the clinical entity and demography, it offers quite a long
period of protection – even up to 50 years [27–29]. A thorough
2.1.5. Hepatitis-A inactivated vaccine
analysis found that in certain groups, protection following
Many modeling and anti-HAV kinetic studies have confirmed a baby BCG vaccine could continue up to 15 years; neverthe­
that a high level of seroprotection can be achieved for at least less, the protective efficacy does eventually wane [30].
Table 1. A summary of currently licensed vaccines grouped based on their duration of protection, along with other vital parameters.
Estimated duration of protection
Correlate of Efficacy Duration of
Number Vaccine Vaccine type protection (CoP) (point estimate) Clinical entity protection Comments
(I) Vaccines that provide long duration of protection (>20 years).
1. Measles vaccine Live attenuated parenteral ≥120 miU/ml 83% (95% CI: 76–88) Measles infection Lifelong protection Boosting of primary immune response is seen with exposure to
vaccine and disease after wild virus or after re-vaccination
seroconversion
2. Rubella Live attenuated parenteral Abs ≥10-15IU/ml 80.7%, (95% CI: 73.7– Rubella infection Lifelong protection Rubella virus-specific humoral immune responses wane with time
vaccine 85.8%) after vaccination, but they do so at a moderate rate.
3. Yellow Fever Live attenuated vaccine (17D log10 neutralization ~99% Yellow fever Lifelong protection Some waning after 10 years. Boosters were recommended after
Vaccine strain) index (LNI) of ≥0.7 disease 10 years till recently.
4. Hepatitis-B Inactivated, sub-unit ≥10 mIU/mL Against infection: 89% Hepatitis-B disease Up to 30 years After 3 doses
Against chronic
carriage: 96%
5. Hepatitis- Inactivated vaccine ELISA Ab 20mlU/ml >98% Hepatitis At least 25 years Mathematical modeling suggests that detectable antibodies are
A-inactivated A infection estimated to persist for as long as 60 years for the 2-dose
schedules.
II. Vaccines that provide moderate duration of protection (5–20 years).
1. Oral Polio Vaccine Live attenuated, oral ≥1/8 dilution of 72% (95%CI: 57–82) Paralytic polio Up to 18 years Data limited from the tropical countries
NAbs
2. Inactivated Polio Inactivated ≥1/8 dilution of 89% (95% CI 62%, 97%) Paralytic polio At least 18 years Limited studies from the developing and tropical countries
vaccine NAbs with two doses:
3. Hepatitis-A-live Live attenuated vaccine (H2 ELISA Ab 20mlU/ml 100% Hepatitis At least 15–17 After a single dose of H2 strain based live attenuated vaccine in
attenuated strain) A infection years endemic region.
Protection may persist longer. Trials on longevity of immunity
are still ongoing.
4. Human Inactivated, ND For HPV naive at HPV vaccine- 11–14 years Longitudinal trials are still ongoing. The longest period of
papillomavirus Virus-like particle (VLP) enrollment: 91–100% induced (following full preventive effect for the bivalent, 4-valent, and 9-valent
(HPV) vaccine based (64.6% to 86%; 94.2% seropositivity course) vaccine were 11 years in the Costa Rica trial, 14 years in the
to 100) FUTURE II, and 8 years in the LTFU extension study and the
Scandinavian study, respectively.
5. BCG Live attenuated, ID ND 37% for all forms of Pulmonary & 10–15 years Protection may last longer, may be up to 50–60 years though
(IFNγ-production tuberculosis (TB) in extrapulmonary waning of efficacy does occur.
by CD4 T cells) children below 5 years TB
of age
42% against pulmonary
disease in children
below 3 years of age
6. Diphtheria Toxoid 0.01–0.1 IU/ml 95.5% (95% CI 92%, Diphtheria disease 11 years After primary series and two booster doses
97.4%
7. Tetanus Toxoid 0.1 IU/ml Virtually 100% Tetanus disease 10 years After three primary doses and two boosters
8. Varicella zoster Live attenuated parenteral GP ELISA ≥5 U/ml 98% All chickenpox Around 10 years Several studies have shown that vaccinated people had varicella
vaccine vaccine disease (after single antibodies for at least 10 to 20 years after vaccination. But
dose) these studies were done before the vaccine was widely used
and when infection with wild-type varicella was still very
common.
9. Mumps Live attenuated parenteral ND 75.8% (35.6%, 90.9%) Mumps disease 6–7 years It is estimated that 85% protection is there 6–7 years after 2
vaccine with Urabe strain and doses of the vaccine and decrease to pre-vaccination level by
64.7% (10.6%, 86.0%) 12 years in the absence of natural boosting
for Jeryl-Lynn strain
EXPERT REVIEW OF VACCINES

10. Pneumococcal Inactivated, polysaccharide 0.20–0.35 mcg/ml 80% (95% CI58–90) Pneumococcal More than 6 years Exact duration of protection is not known; marked variation in the
conjugate conjugate, subunit vaccine (ELISA for Abs) disease protection rates of different serotypes
Vaccine
(Continued )
391
392

Table 1. (Continued).
Estimated duration of protection
Correlate of Efficacy Duration of
Number Vaccine Vaccine type protection (CoP) (point estimate) Clinical entity protection Comments
11. Rabies Inactivated, (Cell culture & NAbs: ≥0.5 IU Virtually 100% Rabies infection 9 years Vaccine induced memory B cells appear to persist for life.
embryonated egg-based Effective recall of the immune response to additional doses,
rabies vaccines (CCEEVs) such as for Post-exposure prophylaxis (PEP), are documented
for decades after priming.
12. Herpes Zoster Recombinant subunit ND 97.2% (95% CI, 93.7–99.0; Herpes Zoster 10 years Unlike the live attenuated vaccine, the high efficacy found even
(recombinant) (glycoprotein E) vaccine p<0.001) Herpes Zoster after 10 years of primary vaccination.
vaccine with AS01 adjuvant ophthamicus
system Post herpetic
neuralgia
13. Pertussis-Whole Inactivated, whole cell ND 94% (95%CI: 88–97%) Pertussis infection 4–12 years After primary series and childhood boosters
V. M. VASHISHTHA AND P. KUMAR

cell vaccines
14. Pneumococcal Inactivated, polysaccharide ND 74% (95% CI: 54–85) Pneumococcal 5–8 years The VE in older adults diminishes in the years following
polysaccharide subunit vaccine diseases vaccination and is likely absent after some interval, which may
vaccine (PPSV) be on the order of 5 or more years.
15. H. influenzae type Inactivated, polysaccharide 0.15 mcg/ml 95–100% (95%CI: 55–100) H. influenzae Up to 6 years after Although vaccine effectiveness declined significantly after the
‘b’ conjugate conjugate, subunit vaccine disease a booster in 2nd first year of life (p<.001), it remained high until the sixth year of
vaccine year life
III. Vaccines that provide short duration of protection (<5 years)
1. Meningococcal Quadrivalent Subunit Serum bactericidal 79% (49% to 91%) at Meningococcal ~5 years Waning of Vaccine efficacy varies with serotype and vaccinees’
vaccine vaccine: Bacterial capsular titer of ≥1:8 using <1 year post single Meningitis, age: faster waning in children < 7 years than at older age
oligosaccharides baby rabbit dose meningococcal
conjugated to a carrier complement fulminant
protein (rSBA) septicemia
2. Pertussis-acellular Inactivated, sub-unit vaccine ND 84% (95%CI: 81–87%) Pertussis infection 4–7 years Evidence of waning of immunity beyond 4 years and little-to-no
vaccines & disease protection beyond 7 years from last vaccination
Primary series, followed by two boosters
3. Dengue vaccine Live attenuated ND 62.0% (95% CI, 56.6–66.7) Virologically 5 years in Waning of protection
(CYD-TDV) viral vaccine (NAbs, variable) at 1 year confirmed seropositive Efficacy varies by sero-status and severity of disease
(CYD-TDV-Dengvaxia ) ® dengue
4. Dengue vaccine Live attenuated tetravalent ND 82.2% (95% CI: 87.6– Virologically 4.5 years Waning of protection
(TAK-003) dengue vaccine (TAK-003, (NAbs, variable) 74.5%) at one year confirmed Efficacy varies by sero-status and severity of disease
Qdenga )™ dengue
5. Cholera vaccine, Whole cell killed vaccine ND 67% Cholera 5 years The combined VE after 5 years was estimated to be 65%
bivalent (Vibriocidal Abs)
6. Cholera vaccine, Whole cell killed vaccine ND 85% Cholera 6 month to 5 years The protection lasts only 6 months for children and 2 years in
monovalnt with recombinant (Vibriocidal Abs) adults.
B subunit of cholera toxin
7. Japanese Live attenuated NAbs: 1:10 (PRNT) 98.5% (CI 90.1–99.2%) JE disease 5 years Recent studies from India find showed a decreasing trend of anti-
encephalitis SA 14-14-2 vaccine (after a single JEV specific IgG antibody titers following two doses of SA-14-
vaccine-live dose) 14-2 vaccine. Marked waning in titers noted after even 2 years
of vaccination
8. Japanese Inactivated NAbs: 1:10 (PRNT) 91% (95% CI: 70–97) JE disease 5 years (after 2 −2 dose primary series in adults from non-endemic regions
encephalitis (IXIARO /JESPECT /
® ® primary doses) –Modeling suggests protection may last up to 14 years after
vaccine- JEEV ) ® booster at 15 mo
Inactivated
9. Typhoid conjugate Inactivated, polysaccharide ND 81.6%; (95% CI: 58.8 to Typhoid fever Around 5–7 years Definite waning in anti-Vi IgG antibodies seen after 5 years. To
vaccine conjugate, subunit vaccine 91.8) extend protection, administering a booster dose to children ∼5
years after their primary dose may be considered
(Continued )
Table 1. (Continued).
Estimated duration of protection
Correlate of Efficacy Duration of
Number Vaccine Vaccine type protection (CoP) (point estimate) Clinical entity protection Comments
10. Typhoid Typhoid polysaccharide ND 69% (95% CI = 28–87%) Typhoid fever 3 years To maintain protection, revaccination is recommended every 3
unconjugated vaccines years.
Vi-
polysaccharide
(ViPS)
11. Herpes zoster Live Live attenuated ND Varies with age; 51% Herpes Zoster ~5 years Efficacy is higher and wanes less for hospitalization for HZ and
attenuated (CD+ T cells overall; 69.8% in 50– Herpes Zoster post-herpetic neuralgia than for Herpes Zoster and Herpes
Vaccine Protective) 59 year ag group and ophthamicus Zoster ophthalmicus
18% in >80 years Post herpetic
neuralgia
12. Rotavirus vaccines Live attenuated oral vaccine ND 77% − 90% in developed, Rotavirus diarrhea 1–2 years Vaccine efficacy is lower and wanes more rapidly in high-mortality
(Secretary IgA) industrialized countries settings than in low-mortality settings
51% in developing
countries
13. Influenza vaccines- Inactivated split & sub-unit HI Abs: 1/40=50% 59% (95%CI: 41–71%) Influenza disease 6 months There is marked variation in the VE of different influenza sub-
Inactivated vaccines protection types. The decline in VE is more pronounced for A H3 than AH1
(1/320 in children) viruses.
*
14. SARS-CoV2 vaccine mRNA vaccine (mRNA-1273 ND 94.1% (95%CI: 89.3–96.8% Covid-19 3–6 months Duration of protection varies according to the severity of the
vaccine) (NAbs seems symptomatic disease, type of circulating variant, highest for the WT and
protective) disease lowest for Omicron and later sub-lineages
ND= Not Determined.
HI=hemagglutination inhibition (HI).
*The immune response to influenza is multifaceted and there are probably multiple correlates of protection.
EXPERT REVIEW OF VACCINES
393
394 V. M. VASHISHTHA AND P. KUMAR

Table 2. Factors determining durability of protective immunity.


Parameters Long durability Short durability
Antigen/Vaccine related factors
Antigen form Particulate antigens (virus particles or aggregated proteins) Homologous monomeric/soluble antigens
Antigen type T-cell dependent (Protein) T-cell independent (polysaccharide)
Antigen valence High Low
Adjuvant/s Addition of depot or slow-release formulations Non-adjuvanted inactivated vaccines
presence &
type
Antigenic Low High
diversity
Vaccine platform Live attenuated (replicating antigen), Virus like particle (VLP) All other platforms (Non-replicating antigens)
Antigen dose Higher Lower
Vaccination At least 4–8 weeks between priming doses and 6 months interval between Schedules with shorter interval
schedule primary doses and booster
Route of Intradermal/subcutaneous administration and heterologous prime-boost Homologous, conventional routes of administration
administration routes may facilitate longevity of immunity
Vaccinee related factors
Age at Older children and adults Extremes of age
vaccination
Gender Female gender Male gender
Genetic factors HLA Class II Polymorphism and individual HLA genotype may affect immunity HLA Class II Polymorphism may have a negative impact on
immunity
Gut microbiome Individual’s baseline gut microbiome composition and gut metabolome may Individual’s baseline gut microbiome composition and gut
positively impact immunity metabolome may have negative impact on immunity
Target pathogen and disease related factors
Incubation period Long Short
Force of infection Lower High
Site of infection Systemic infection Mucosal infection
Clinical endpoint Protection against severe disease Protection against infection

2.2.6. Diphtheria have demonstrated a waning in immunity after a single dose of


Protective levels of diphtheria antibodies (0.1IU/mL) persist for the OKA-strain varicella vaccine [37]. Though anti-varicella anti­
decades after vaccination, although they wane in a section of bodies are estimated to have a half-life of 50 years, there are
vaccinees after ten years [31]. The half-life of diphtheria-specific frequent fluctuations in the titers.
antibodies is estimated to be around 19 years [32]. Considering
this and the fact that diphtheria infection has a short incubation 2.2.9. Mumps vaccines
period, and the natural boosting is also limited with high vacci­ Although wild mumps virus infection provides lifelong immu­
nation coverage, its booster is recommended every ten years, nity, recurrences of mumps disease are reported [38]. There
especially in industrialized countries, to maintain protective are at least 13 different strains of Mumps virus employed in
levels. However, recent studies have suggested that the protec­ live attenuated vaccines [39]. They differ in efficacy to a certain
tive titers persist much longer in the majority, and less frequent extent, but overall, they protect 80% of the recipients.
boosters should be sufficient [33]. Some studies have reported 60–90% effectiveness after
a single dose [39]. According to a Cochrane review of 64 MMR
2.2.7. Tetanus vaccine studies, one dose’s effectiveness was 69%-81% [40].
The duration of protection accorded by the tetanus vaccine is also Another study finds that mumps-specific antibody titers per­
similar to the diphtheria antigen. The protective level of 0.1 IU/ml sist for at least 32 months after vaccination and slowly wane off. It
of anti-tetanus antibodies persists for 3–5 years after the primary is estimated that 85% protection is there 6–7 years after two
schedule of 3 doses. Then, it may persist lifelong after boosters in doses of the vaccine [41] and decreases to pre-vaccination level
the second and fifth years of life and another during the adoles­ by 12 years without natural boosting [42]. The antibody titers
cent age in some people [33]. The half-life of tetanus-specific persist longer with natural boosting: for example, a study from
antibodies is estimated to be around 11 years [31]. However, in Finland reported 74% seropositivity 20 years after MMR vaccina­
most persons, the level approaches minimal protective levels ten tion [43]. A longitudinal study estimated the half-life of anti-
years after the last dose [34]. Thus, its booster is recommended mumps antibodies to more than 500 years (95% CI: 90 to infinity)
every ten years to maintain protective levels. Recent studies have and showed no significant decrease in titers over time [3].
indicated that less frequent boosting might be enough [35].
2.2.10. Pneumococcal conjugate vaccines (PCVs)
2.2.8. Varicella zoster vaccine Though the exact duration of protection has yet to be
Many studies from the U.S.A. and Japan demonstrated the long- established, PCVs provide reasonably good protection fol­
term persistence of anti-varicella antibodies after a single dose lowing a primary infantile schedule [44]. There are few long­
of varicella vaccine [36]. However, there are concerns that these itudinal studies to measure persistent immunity against
studies were conducted when the virus had a substantial circu­ pneumococcal diseases. Different experts have observed
lation in the community, and there may be some waning now a marked variation in the protection rate of different sero­
due to the absence of widespread transmission. Later studies types. A study with PCV7 on nasopharyngeal carriage
EXPERT REVIEW OF VACCINES 395

noticed sustained protection for five years [45]. In South 2.2.15. Haemophilus influenzae type b (hib) conjugate
Africa, an experimental PCV, PCV9, showed consistent pro­ vaccine
tection for 6.3 years after vaccination against Invasive Haemophilus influenzae type b (Hib) conjugate vaccine is
Pneumococcal Disease (IPD) [46]. highly immunogenic with an estimated efficacy of 95–100%
after primary series [60]. Most experts believe that Hib con­
jugate vaccine provides a long-lasting immunity despite
2.2.11. Rabies vaccine waning in titers [61]. However, no exact duration of protec­
Vaccine-induced neutralizing antibody (VNA) after rabies vacci­ tive immunity is known, and the need for a booster dose
nation is considered to be a valuable correlate of protection following the primary schedule is still uncertain [62]. It is
against rabies, and a titer of ≥ 0.5 IU is taken as a cutoff [47]. generally believed that the Hib conjugate vaccine protects at
Briggs et al. found that 80% of vaccinated subjects still had least for five years after completion of the primary series.
detectable VNA titer after nine years [47]. Significantly, they did Many countries have successfully controlled the Hib disease
not find any correlation between the VNA titer and the number with a three-dose primary schedule, but some countries had
of vaccine doses received as a part of PrEP or PEP or the time to introduce a booster dose after a few years [62,63]. As per
elapsed since the last dose of the vaccine series. However, the some studies, the protection offered by three doses in
intramuscular (IM) schedule induced neutralizing antibodies for infancy lasts only till 2nd year of life and wanes quickly
a longer duration than the intradermal schedule [48]. A German [64,65]. The anamnestic response to booster dose is excel­
study of the longevity of vaccination-induced rabies VNA found lent [64].
detectable antibodies in 18 subjects 14 years after their last dose
in the Pr-EP and PEP schedules [49]. Furthermore, rabies vaccine
induced a robust immune memory that provides excellent
‘immune recall’ even after several years of primary schedule [50]. 2.3. Vaccines that provide short duration of protection
(<5 years)

2.2.12. Herpes zoster recombinant vaccine 2.3.1. Meningococcal vaccines


This new zoster vaccine is based on glycoprotein E of the Meningococcal polysaccharide vaccines, being T-cell indepen­
varicella-zoster virus (VZV) and has an AS01B adjuvant system. dent vaccines, do not provide durable protection/memory-cell
The persistence of humoral immune response [anti- response. In younger children, 2–4 years of age, the protection
Glycoprotein E (gE) antibodies] and cellular immune response lasts only a few months, while in older children, it lasts 2–3
(gE-specific CD4+ T-cells) till six years after two doses of the years. Revaccination again protects for a similar duration
vaccine has been demonstrated [51]. Boutry et al. followed up [66,67].
with 7413 participants enrolled in the long-term follow-up Meningococcal conjugate vaccines offer better protection
than polysaccharide vaccines, but even with these, the effi­
study of the previous clinical trials to study the durability of
cacy wanes over the years. The serological correlate of pro­
vaccine efficacy. They found 84.1% efficacy after eight years of
tection is serum bactericidal titer of ≥ 1:8 using baby rabbit
the second dose [52].
complement (rSBA) [68,69]. Immunogenicity study in children
aged 7–9 years has shown that seropositivity rates fall from
2.2.13. Whole cell pertussis (wP) 97.3% to 63.6% for serogroup A and 87.9% for other ser­
Various studies have estimated the duration of protection ogroups (C, W, and Y135) 2 years after vaccination and that
offered by whole-cell pertussis (wP) vaccines, which ranges falls to a mere 20% after six years [70]. Cohn et al. have
from 4 to 12 years [53]. Long-term studies have shown shown that the vaccine efficacy of a single dose falls from
a definite waning of protection after wP vaccines and natural 79% (49% to 91%) at <1 year post-vaccination to 61% (25%
infection over this period [53–55]. A study by Campbell et al. to 79%) at 3 to <8 years post-vaccination [71]. The waning in
from the UK cited 83.7% efficacy in children 10–16 years of age the protective antibody titers following meningococcal con­
following three primary doses, though a decline in vaccine jugate vaccines also varies with the child’s age, with waning
efficacy was noticed within 1–2 years of the third dose [56]. faster in kids under five years of age than kids above five
According to a systematic review, annual loss in the efficacy of years of age [70].
wP vaccines ranges from 2–13% [57].
2.3.2. Acellular pertussis (aP) vaccines
2.2.14. Pneumococcal polysaccharide vaccine (PPSV) Compared to wP vaccines, waning is faster with acellular
Pneumococcal polysaccharide vaccine is recommended in older pertussis (aP) vaccines. Schwartz et al. observed that the dura­
adults and children with specific risk factors. On vaccination, the tion of protection following aP vaccines declined rapidly fol­
antibody levels decline over time after an initial rise and reach lowing four years of vaccination despite high early
almost baseline levels within 4–7 years of vaccination [58]. effectiveness [72]. The efficacy drops from around 90% in the
Extensive studies in older adults have shown vaccine effective­ first year after the last dose of the aP vaccine to 30–40% 6–8
ness wanes over time, and effectiveness may be absent after five years after the last dose [73–76]. Waning of protection after
years. However, a large cohort study, mainly in adults over 65, TdaP vaccines is even faster: some studies have shown the
has shown good protection against severe pneumococcal infec­ protection wanes quickly, with efficacy dropping to as low as
tions even after nine years [59]. 8.9% after four years [77–80].
396 V. M. VASHISHTHA AND P. KUMAR

2.3.3. Dengue vaccines the persistence of sero-protection for six years after two pri­
Currently, two live attenuated dengue vaccines – CYD-TDV mary doses of IXIARO and a booster dose after 15 months [91].
(Dengvaxia™ by Sanofi) and TAK-003 (Qdenga™ by Takeda)
are licensed and approved for use in different populations in
2.3.6. Typhoid vaccines
some countries. According to data from three efficacy trials,
Vi-polysaccharide vaccine: A T-cell independent antigen, it
CYD-TDV offers protection for five years against severe dengue
does not induce memory cells and immunoglobulin class
among seropositive individuals [81]. Another live dengue vac­
switching. Hence, it does not offer long-term protection and
cine, TAK-003, has been evaluated for 54 months. The pub­
requires revaccination every three years for continued pro­
lished data showed a 62.0% efficacy against virologically
tection [92]. Even within the three years, vaccine efficacy
confirmed dengue (VCD) and 83.6% against hospitalized VCD
falls from 64% after 21 months to ~ 55% in the
[82]. According to unpublished follow-up data, the vaccine
third year [93].
offered 64.2% vaccine efficacy against VCD in seropositive
Typhoid conjugate vaccines: Conjugate vaccines, being
individuals and 53.5% VE in seronegative individuals after 54
T-cell dependent, induce robust immune response with the
months. The vaccine efficacy against hospitalized dengue was
production of memory cells, thus offering long-term protec­
more than 80% after 54 months of follow-up [83].
tion. One study conducted with the vaccine developed with
a nontoxic recombinant protein that is antigenically identical
2.3.4. Cholera vaccines to Pseudomonas aeruginosa (Vi-rEPA conjugate vaccine)
Two types of cholera vaccines are available globally: monova­ among two to five-year-old Vietnamese children demon­
lent and bi-valent-killed whole-cell oral cholera vaccines. While strated 89% efficacy after 27 weeks of active surveillance fol­
the prototype of the former is WC-rBS (recombinant B subunit lowed by 19 weeks of passive surveillance [94]. Studies
of cholera toxin – Dukoral®), there are three different bi-valent performed on another conjugate vaccine using tetanus toxoid
vaccine products available globally that contain O1 and O139 as a carrier protein, Vi-TT (Typbar-TCV™), demonstrated that
strains (without the B subunit) and marketed as Shanchol™, protection following a single dose might persist for up to five
Euvichol™ and mORCVAX™ All of them, barring mORCVAX™ years [95,96]. A longitudinal study finds the persistence of
(mainly used in Vietnam), are WHO-Pre-qualified [84]. anti-Vi-IgG antibodies up to 7 years following primary vaccina­
The estimated duration of protection of these cholera vac­ tion of children in both boosted and un-boosted cohorts.
cines ranges from 6 months to 5 years. For the monovalent However, there was a significant waning in the titers after
Dukoral™ vaccine, the protection lasts only six months for five years, and the manufacturer recommended a booster
children and two years for adults. For the bivalent, whole-cell after five years of the primary dose [96].
vaccine, Shanchol™, protection lasts up to 5 years after vacci­
nation. In a large RCT conducted in Kolkata, the efficacy dif­
fered in different age groups – highest in older children and 2.3.7. Herpes zoster live attenuated vaccine
adults and lowest in young kids. The combined VE after five This live attenuated vaccine gives protection for a shorter
years was estimated to be 65% [85]. duration than the recombinant, inactivated vaccine. Efficacy
against Herpes zoster and post-herpetic neuralgia hospitaliza­
tion is higher and better maintained. A real-world effective­
2.3.5. Japanese encephalitis vaccines ness study demonstrated that herpes zoster vaccine efficiency
Japanese encephalitis vaccine-live (SA-14-14-2): Studies from decreased from 67% (95% CI: 65% to 69%) in the first year to
Nepal show that this live attenuated JE vaccine based on the 15% (5% to 24%) after ten years [97]. Efficacy against post-
SA-14-14-2 strain provides reasonably good protection for five herpetic neuralgia dropped from 83% (78% to 87%) to 41%
years after a single dose. A case-control study in Nepalese chil­ (17% to 59%) during the same period. Vaccine efficacy against
dren found 96.2% effectiveness after five years of a single dose herpes zoster ophthalmicus decreased from 71% (63% to 76%)
[86]. Another case-control study among adults finds 77.0% (95% to 29% (18% to 39%), while that against herpes zoster hospi­
CI: 67.0–83.0) effectiveness after seven years of a single dose
talization dropped from 90% (67% to 97%) to 53% (25% to
administration in two districts of Assam, India [87]. However,
70%) in five to eight years [97].
a recent cross-sectional study conducted in the Gorakhpur dis­
trict of India finds that 97.74% of samples were negative for anti-
JEV-specific IgG antibodies after two years of the second vaccine 2.3.8. Rotavirus vaccines
dose [88]. Vaccine effectiveness of live attenuated rotavirus vaccines
Inactivated Vero cell-derived vaccines (IXIARO®/JESPECT® declines more rapidly in regions with high burden-high mortality
(Valneva, Austria) & JEEV®(BE, India): Data on the long-term than in industrialized countries having low burden-low morbidity
protection against the clinical disease from the endemic [98]. According to a meta-regression study, the efficacy of live
region is limited. A Philippine study with IXIARO® among oral rotavirus vaccines waned more rapidly in high-mortality
300 children finds that 90% of un-boosted subjects had pro­ settings, and pooled efficacy after 12 months was only 44%. In
tective anti-JE antibodies after three years of primary vaccina­ contrast, in low and middle mortality settings, the corresponding
tion of two doses [89]. A 5-year follow-up study among two- figure was 77%–94% [99]. Some efficacy studies from the low
dose recipients of the IXIARO vaccine in Austria finds that 81% burden-low low mortality countries have demonstrated sus­
of the subjects retained the protective antibody titers [90]. tained protection against severe rotavirus disease even after
Another study from the non-endemic region demonstrated three years of primary vaccination [100,101].
EXPERT REVIEW OF VACCINES 397

2.3.9. Influenza vaccines Another review based on 41 studies on the performance of


Vaccines against influenza do not produce long-term protec­ exclusively inactivated COVID-19 vaccines found that primary
tion. After immunization, the anti-HA antibodies peak 2–4 VE had waned significantly after six months [110]. The study
weeks after vaccination in primed individuals, whereas in noted VE of 52.8% against the Delta variant, which went down
unprimed individuals and older adults, they peak ≥4 weeks sharply against the Omicron (16.4%). However, like mRNA
after vaccination. After six months, the titers drop by more vaccines, the VE against severe disease remained good even
than 50%, with the amount of the drop proportional to the after six months of primary series, which improved after
highest titers attained. The titers then stay constant for two to a booster dose [110].
three years [102]. According to clinical trial data, immunity
against antigenically identical strains to those in the vaccina­
3. Immunological basis of long-term protection
tion lasts for roughly 6–8 months [103].
conferred by vaccines
The vaccine effectiveness (VE) of the various influenza sub­
types varies significantly. AH3 viruses have a more marked Vaccines induce immune responses that are highly complex
decrease in VE than AH1 viruses. Moreover, the VE is influ­ and include orchestrated actions of a wide range of immune
enced by the interval since immunization. According to cells in both innate and adaptive immune systems (Figure 1).
a comprehensive review and meta-analysis, VE for influenza Ultimately, with most current vaccines, protection is provided
virus subtypes A/H3 and type B is significantly less at 3–6 through pathogen-specific antibodies, which target specific
months after vaccination than 0–3 months after vaccina­ epitopes in pathogens or their toxic components. Antibodies
tion [104]. are exclusively produced by a subset of B-lymphocytes
A different study discovered that the influenza A(H3N2), (B-cells) called plasma cells. The process of antibody produc­
influenza B, and influenza A(H1N1) pdm09 viruses caused tion begins with the vaccine antigens reaching the draining
a 7% (absolute) monthly drop in VE. Patients who had lymph nodes from the injection site, where they are captured
received influenza vaccinations in the previous season showed by specialized macrophages in the sub-capsular region and
a more marked decline in VE [105]. An Australian study dis­ then transported to B- -cell zone (follicles) (step ‘a’ in Figure 1).
covered an intra-seasonal drop in influenza vaccine effective­ Here, B-cells are activated, and activated B cells migrate
ness. The study found that the best defense against influenza toward the border region between B-cell follicles and the
verified in a lab was when immunization was given within two T-cell zone (called marginal zone) [111].
months of the onset of symptoms [106]. Vaccine effectiveness After that, there are two pathways: One, activated B-cells
could disappear as soon as 90 days after inoculation, as per can induce extra-follicular response (without any help from
a 2018 analysis of 11 recent studies on the vaccine’s durabil­ T-cells), which leads to the production of short-lived plasma
ity [107]. cells (step ‘b’ in Figure-1). Bacterial antigens like lipopolysac­
charides induce this type of response. As the name suggests,
these short-lived plasma cells secrete antibodies for a short
2.3.10. SARS-CoV-2 vaccines time; thus, the vaccine-induced protection is only short dura­
There is no known broad estimate of the duration of protec­ tion. Without T-cell help, the antibodies produced are of low
tion offered by the SARS-CoV-2 vaccines against COVID-19 to modest affinity and mostly IgM type with no isotype switch­
disease, which varies according not only to the disease state ing [111]. Of note, this reaction is relatively rapid, leading to
and type but also to the circulating variants. Further, despite the appearance of IgM antibodies within a few days of vacci­
the extensive use of different vaccines and hundreds of stu­ nation. Vi Polysaccharide typhoid vaccine is a classic example
dies over three years, no reliable immune correlate of protec­ of this [112].
tion against the disease could be ascertained so far. Overall, The second pathway is the germinal center (GC) response
neither the infection nor the vaccines provide lasting protec­ induction. This pathway involves intricately coordinated activ­
tion. The effectiveness of these vaccines rapidly wanes over ities in different lymph node zones and is relatively slower: it
time after primary and booster vaccination. Most studies, takes roughly two weeks for the germinal centers to develop
including systematic reviews, found that protection waned [113]. In the T-cell zone, the antigen is presented by major
significantly within 3–6 months of a vaccine dose, especially histocompatibility complex (MHC) class II of antigen-
against the Omicron variant. presenting cells (APC) like dendritic cells (DC) to naïve CD4+
An extensive review evaluated 78 studies on vaccine- T cells, leading to their functional differentiation to helper
specific vaccine efficacy/effectiveness of four COVID-19 vac­ T cells (Th cells) (step ‘c’ in Figure-1). These Th cells provide
cines before the advent of the Omicron variant concluded that helper signals to activate B-cells in the marginal zone to lead
VE against symptomatic disease waned by 20–30% by six to extra-follicular response, producing short-lived plasma cells
months of the primary series. However, the VE remained (step ‘d’ in Figure 1). These short-lasting plasma cells start
high, around 70% against severe disease, and declined only dying after some time, resulting in reduced antibody titers
10% after six months [108]. noted 4–8 weeks after vaccination [114]. However, because
Another review based on 40 peer-reviewed studies finds T-cells help in the form of type-1 cytokine interferon-gamma
that the VE against the omicron variant was only 20% after six (IFN-γ) (produced by follicular helper T-cells (TFH cells) and
months of the primary series and 30% after nine months of Th1 cells) or Type 2 cytokines interleukin-4 (IL-4) [produced by
a booster dose – a faster waning of VE noted against the TFH and Th2 cells], there is isotype switching to IgG1/IgG3 or
Omicron than the Delta variant [109]. IgG1 and IgE respectively. Some primed CD4+T-cells are
398 V. M. VASHISHTHA AND P. KUMAR

Figure 1. Schematic representation of vaccine-induced immune responses in the draining lymph node.
(Adapted from: Chen Z, Gao X, Yu D. Longevity of vaccine protection: Immunological mechanism, assessment methods, and improving strategy. View, 2022; 3,
20200103).

committed to differentiation toward TFH cells (step ‘e’ in differentiate into plasma cells, secreting large amounts of
Figure 1), and they support the initiation and maintenance high-affinity antibodies (secondary immune response) [115].
of GC reaction [114,115]. The decline of antibody titers after this secondary immune
The GC reaction is initiated by the migration of some response is much slower than that of the primary immune
activated B cells toward germinal centers on getting signals response. Thus, a rapid increase in antibody titer is noted
from primed T-cells and follicular dendritic cells (FDCs) (step ‘f’ within days of infection with the same pathogen later or
in Figure 1). The FDCs are essential here: they attract antigen- after a booster dose of the vaccine. During the GC reaction,
specific B and TFH cells and capture/retain antigens for a fraction of plasma cells can migrate toward long-term survi­
extended periods. B cells attracted by antigen-bearing FDCs val niches mainly located within the bone marrow, where their
become the founders of the GC reaction. Each GC contains survival and antibody production may persist for years [116].
B-cells of a single antigen only. These B-cells undergo massive The long persistence of antibodies can be predicted by mea­
clonal proliferation, somatic hypermutation, and affinity suring antibody titers between 6–12 months of vaccination, as
maturation [selection of fittest B-cells (with highest affinity) by this time, all short-term plasma cells have disappeared, and
after several ‘rounds’ of selection]. in the absence of repeat vaccination, the antibodies in serum
Moreover, they differentiate into long-lasting plasma cells are those secreted by long-lasting plasma cells only [117].
(step ‘g’ in Figure 1) that secrete large amounts of high-affinity In addition to this antibody-mediated immune response
antibodies for extended periods (primary immune response) (humoral immune response), CD8+ T cell-mediated cytolysis
[115]. Some GC B-cells differentiate into memory B cells (step is a crucial mechanism of cellular immunity against intracellu­
‘h’ in Figure 1). Memory cells persist as resting cells until re- lar infections. It is induced in the T-cell zone where naïve CD8+
exposed to the same antigen when they re-proliferate and T-cells are activated by cross-presentation of the antigen via
EXPERT REVIEW OF VACCINES 399

the MHC class I molecules on dendritic cells that drive their not exist for all vaccines in real life. While in the case of
differentiation into short-lived effector CD8+ T-cells (step ‘i’ in specific antigens like measles and rubella, the memory B-cell
Figure 1) or resident memory CD8+ T-cells (step ‘j’ in Figure 1) population remains stable and correlates well with antibody
[118]. Like a humoral immune response, most short-lived levels decades later, whereas, in the case of some other anti­
cytotoxic effector cells enter programmed cell death at the gens like varicella – zoster virus, Epstein – Barr virus, tetanus,
end of primary immune responses. In contrast, memory CD8+ or diphtheria, no such correlation exists [3]. It is also noticed
T cells persist over a more extended period, and they can that this association fails to exist for those vaccines requiring
rapidly proliferate into a massive wave of effector cells upon multiple or repeated doses. This observation reflects that the
re-exposure to the same pathogen. In addition to protecting mere persistence of memory B-cells may not guarantee the
intracellular antigens, the memory function of CD8+ T cells has durability of antibodies, and some other mechanism may be
been suggested to be a significant protection mechanism for involved in maintaining the long-term antibody levels. One
the elderly with inadequate antibody responses [114]. such mechanism could be ‘imprinted lifespan’ phenomenon in
Moreover, a new subset of CD8+ T cells that persist in non- which B-cell signaling during the induction of antigen-specific
lymphoid tissues for an extended period and can control immune response determines the life span of the plasma cells
reinfection has been recently identified. Termed tissue- [121]. Most antigens induce memory B-cell formation, but the
resident memory T cells (TRM) they are of great interest for duration of antibody production is not the same. This hypoth­
developing vaccines against infections entering through esis may explain why certain antigens provide long-term
respiratory and digestive tracts where conventional antibody- immunity while others provide only short-term protection.
based vaccinations cannot induce sufficient capacity to neu­ During the process, the exact mechanisms responsible for
tralize pathogens during initial infections [119]. differentiating immune cells toward different subsets and
The critical determinants of the persistence of humoral pathways are also poorly understood even today. However,
immunity are two broad immune mechanisms – memory the induction of good GC reactions, many long-lasting plasma
B-cell dependent and memory B-cell independent. The immu­ cells, and the formation of memory B-cells and T-cells are
nity in the latter is provided through antibodies produced by essential for durable, long-lasting vaccine-induced protection
the long-lived plasma cells residing in the bone marrow niches [115] (Figure 2).
[120]. The memory B-cells themselves do not provide protec­
tion, but they divide and differentiate into plasma cells after
antigenic exposure to secrete large amounts of antigen-
4. Determinants of the longevity of antibody
specific antibodies [3]. So, to assess the longevity of immunity
response
induced by an immunogen, measuring the memory B-cell
counts should provide an estimate of the durability of immu­ Certain known factors determine how long the vaccine-
nity. However, studies have shown that this correlation does induced antibody responses can be expected to last (Table 2):

B-Cells
Spleen

c . Long lived plasma cells


b. Memory B cells
a . Germinal centers in a lymph node

T-Cells

Thymus

d. Memory T cells
e. Tissue resident memory T cells

Figure 2. Key immunological determinants of durable vaccine-induced immune responses.


Both B-cell and T-cell responses help in providing durability to vaccine-induced immune responses. A. Germinal centers (encircled) in lymph nodes are factories of
B-cell production and their maturation. B. Memory B cells in lymphoid tissues like the spleen. C. Long-lived plasma cells move to the bone marrow niches and
survive there, providing long-lasting immunity.
D. Memory T-cells in the lamina propria and epithelium of the intestine. E. Tissue-resident memory T-cells (TRM) in the nasal passages and lungs. These cells are in
non-lymphoid tissues, such as skin, gut, and lung airways, and do not recirculate through the blood. Most T cells mature in the thymus gland before subsequent
export to the periphery.
400 V. M. VASHISHTHA AND P. KUMAR

4.1. Vaccine- and vaccination-related factors this principle. For instance, the recently approved RTS,
S vaccine for malaria consists of a repeating, particulate anti­
4.1.1. Vaccine platform
gen combined with a potent adjuvant [127]. Nevertheless, this
The vaccine platform is crucial to determining longevity.
vaccine induces a protective immune response only in around
Among all vaccination platforms, only live-attenuated and
36% of individuals at vaccination, and the level of protection
virus-like particle (VLP)-based vaccines elicit long-lasting
diminishes to 0% over three years [128].
antibody responses that can endure for several decades,
Another recent development in vaccinology is the ‘struc­
or even a lifetime, without the need for further exposure
ture-based’ design of protein antigens (now termed ‘designer
to the antigen or reactivation of immunological memory.
antigens’), which delineate the antigenic structures in the ‘pre-
Amanna et al. conducted a long-term study on the levels of
fusion’ and ‘post-fusion’ forms. The advent of new imaging
specific antibodies against viral antigens (vaccinia, measles,
techniques like X-ray crystallography and cryogenic electron
mumps, rubella, varicella-zoster virus, and Epstein-Barr
microscopy (Cryo-EM) has made it possible to study these
virus) and non-replicating antigens (tetanus and diphtheria)
forms. Antibodies directed against the ‘post-fusion’ (after the
in 45 individuals over up to 26 years. They discovered that
antigen fuses with the host cell) shape do not thoroughly
the antibody responses against the viruses remained
neutralize the circulating form of the virus before it binds to
remarkably stable, with estimated half-lives ranging from
cells, known as the ‘prefusion’ form. Antibodies directed
50 years for the varicella-zoster virus to over 200 years for
against the epitopes, exposed during only the pre-fusion
other viruses like measles and mumps [3]. The antibody
form, are highly potent in neutralizing the virus. The knowl­
responses to non-replicating antigens (tetanus and
edge about this concept led to the successful development of
diphtheria) declined faster, with estimated half-lives of 11
respiratory syncytial virus (RSV) vaccines and has also been
years and 19 years, respectively [3,121]. Live vaccinations, in
employed in designing a few mRNA and protein-based COVID-
contrast to non-live antigens, cause activation of several
19 vaccines [129].
lymph nodes due to the multiplication and spread of
microbes [117]. The process of microbial replication leads
(1) Antigen type is a crucial determinant of the longevity of
to a constant production of antigens, which in turn con­
immune responses elicited by a vaccine. Polysaccharide
tributes to an intense and persistent immune response.
antigens are ‘T-cell independent’ and do not induce
VLP-based vaccines, such as the Human Papilloma Virus
TFH cells and GC reaction in the absence of T-cell
(HPV) vaccine, consist of supramolecular nanoparticles with
help (vide supra) [117]. Thus, the antibody response to
highly repetitive antigen sequences displayed on their sur­
these antigens is predominantly IgM type, low titers of
faces. The high antigen valency of VLPs, which refers to
low-affinity IgG, and only of short-duration [117].
how densely the vaccine antigen is presented to the
Hence, the antibody response’s durability and ‘quality’
immune system, can significantly enhance early B-cell acti­
are suboptimal. Conjugation of polysaccharide antigen
vation and the persistence of GC responses. This, in turn,
to a ‘carrier’ protein can lead to induction of TFH cells,
leads to increased, long-lasting plasma cell differentiation
GC reaction, and thus high-affinity antibody response
and thus results in a durable and long-standing immune
with isotype switching and immune memory, leading
response [122].
to longer-term protection, as seen with Pneumococcal
COVID-19 vaccines are attractive, as multiple vaccines have
conjugate and typhoid conjugate vaccines [130].
been developed, primarily based on its spike antigen, but on
(2) Adjuvants. Adjuvants improve the immune response to
different platforms [123]. For example, Moderna’s mRNA vac­
inactivated vaccines in many ways, such as modifying
cine induces high titers of vaccine-induced binding and neu­
how the antigen is delivered, how long it persists (using
tralizing antibodies and CD4 and CD8 cells. However, these
depot or slow-release formulations), and boosting Th
wane off by six months, while Janssen’s vaccine, which is
responses (immunomodulation) [131]. Adjuvants can
a virus vector (Adenovirus-26) based vaccine, induces anti­
enhance the durability of vaccine-induced immunity
body titers, CD4 and CD8 cells that have lower peak than
by enhancing both the quality and intensity of immu­
mRNA vaccine, but persist longer (up to 8 months) [124].
nological responses.
Further, studies have shown that boosting with a vaccine
(3) Antigen dose is also a factor as far as the durability of
from a different platform (heterologous boosting) induces
vaccine-induced immunity is concerned. An increase in
a better immune response than a homologous schedule [125].
antigen dose, in general, enhances immune response
by increasing the availability of the antigen for B/T-cell
4.1.2. Antigen form binding. However, there is some complexity as the
The form of the antigen influences the immune response. antigen dose is also shown to impact the induction of
Particulate antigens, such as virus particles or aggregated T-regulatory cells (Tregs) apart from affecting Th1/Th2
proteins, stimulate more effective antigen uptake and migra­ bias and recruitment of Th17 and TFH cells [132].
tion by dendritic cells. They lead to more robust responses (4) The vaccination schedule also regulates the magnitude
from CD4+ and CD8+ T cells than when the antigen is mono­ and duration of the antibody response. When priming
meric or soluble [126]. Therefore, particle antigens and heat- doses are administered at intervals of less than one
or chemical-induced aggregated antigens elicit a more robust month, the resulting immune responses are less long-
and long-lasting immune response than soluble, monomeric lasting than when the same number of vaccine doses
antigens. Nevertheless, several practical instances deviate from are given at longer intervals (1-2 months). This is
EXPERT REVIEW OF VACCINES 401

because fewer post-GC B cells are generated, which are molecules [human leukocyte antigen A2 (HLA A2)] affect T-cell
capable of long-term survival. A shorter period between responses significantly [138].
waves of primary reactions increases competition, Disease conditions: Certain disease conditions also affect
negatively impacting durability. For the best memory vaccine antibody persistence. Increased catabolism (as in HIV)
retrieval and enhanced immune responses, it is neces­ or urinary or digestive tract antibody loss reduces vaccination
sary to have longer intervals of at least 3 to 4 months. antibody persistence. Immunosuppressive medicines and con­
The optimal timing for a booster dosage is at least 4-6 ditions impact the durability of vaccine-induced protection.
months after the last priming dose, allowing for the Recent studies suggest obesity may accelerate the waning of
necessary affinity maturation of B-cells in the germinal COVID-19 and influenza vaccine efficacy [139].
center and creating memory cells [117]. Administering Gut microbiome : Recent research reveals that the gut
the booster dose ‘prematurely’ leads to elevated anti­ microbiome impacts the development of the immune system
bodies at the boosting time, creating antigen-antibody and immunological response to vaccines. The way by which
complexes, which diminishes the antigens available for the establishment of gut microbiome at/after birth affects
B-cell priming. Additionally, antibody-mediated feed­ childhood vaccination response is unknown [140,141].
back mechanisms may directly affect B cells negatively.
The optimal schedule for non-live T-cell dependent
4.3. Target-pathogen related factors
immunizations that operate on the ‘prime-boost princi­
ple’ is ‘0, 1, 6 months’ [117]. The target pathogen and its pathogenicity significantly
(5) Route of administration. The immunological response to influence the immune response and longevity. For instance,
a vaccine can differ depending on the route of admin­ the incubation period and the force of infection transmis­
istration. For instance, Styles et al. showed in their sion (the pace at which illness is acquired in a community)
animal study that intradermal vaccination led to better impact the effectiveness of vaccines. A lengthier incubation
antigen retention in the lymph nodes than intramuscu­ period allows for a more efficient memory response and
lar administration, which resulted in a more robust and enhanced long-term protection against symptomatic illness
longer-lasting activation of antigen-specific GC T and [124]. This phenomenon has been extensively documented
B cells [133]. Jegaskanda et al. showed that intranasal during the SARS-CoV-2 pandemic, wherein the level of pro­
live-attenuated influenza vaccine, followed by a booster tection against infection declined as the incubation periods
of intramuscular subunit vaccine, resulted in a robust of new strains of SARS-CoV-2 reduced [142]. Moreover, it
immune response [134]. Similarly, administering intra­ has been found that the effectiveness of vaccines is gen­
muscular mRNA-lipid nanoparticles as the initial dose, erally greater in regions where viral transmission, or the
followed by intranasal protein vaccine as a booster, led force of infection, is lower. This phenomenon was demon­
to a strong and effective immune response against strated in the case of a pentavalent rotavirus vaccine, which
SARS-CoV2 [124]. Hence, a combination of various plat­ showed real-world efficiency ranging from 45% to 90%
forms and delivery methods can enhance the durability [143]. OPV also exhibited significant variations in effective­
of protection. ness [144].
Licensed and experimental vaccines against viruses that
replicate on mucosal surfaces without causing systemic
infection/significant viremia also fail to induce durable pro­
tection. Natural infection with these viruses [like influenza,
4.2. Vaccinee-related factors
parainfluenza, RSV, SARS-CoV2, and other ‘human common
Age at vaccination: The vaccinee’s age at the time of vaccina­ cold viruses’] also does not provide long-term protection.
tion also affects vaccine antibody persistence, as the response The possible factors include: (a) They do not come in con­
is shorter at both extremes of age because of immaturity and tact with multiple immune compartments and immune-
senescence of the immune system, respectively [117]. In competent cells, unlike other viruses like measles, mumps,
infancy, the immune response to live attenuated vaccines is rubella, smallpox, etc., (b) they have a shorter incubation
also adversely affected by preexisting antibodies transferred period, as they start causing illness just after mucosal repli­
antenatally through the placenta [135]. Even immune cation (the time required for systemic spread after mucosal
response to non-live vaccines is affected by these maternally- replication is cut-off) [1]. Due to the shorter incubation
derived antibodies but to a lesser extent. period, the immune system does not get adequate time
Gender: Although this aspect is not yet well studied, some to mount an adequate/robust immune response; (c) the
indications indicate that immune response might also vary half-life of nasal secretary IgA is much shorter than serum
with gender. Studies have demonstrated that females elicit IgG. Thus, the viruses that induce mainly mucosal immune
more exuberant immune responses to infections than males responses do not result in durable protection against these
[136,137]. viruses [145,146]. There is another explanation from an
Genetic factors: Antigen epitopes associated with many evolutionary point of view: Unlike other immune compart­
MHC molecules are more likely to induce response. Gene ments, mucosal surfaces regularly come in contact with
polymorphisms in critical molecules required for B and T cell a much larger number of antigens. Hence, they have
activation/differentiation molecules affect the quality of anti­ evolved to ‘tolerate’ less dangerous antigens and down-
body responses. Individual and community variations of MHC regulate immune responses to them to minimize bystander
402 V. M. VASHISHTHA AND P. KUMAR

tissue damage, focussing on more dangerous antigens for [153]. Prolonging vaccine delivery also improved the quality
robust immune response. The mechanisms for this immune of antibody response, as shown by significantly high levels of
‘decision-making’ have not been fully understood yet [1]. high avidity, balanced IgG subclass, and cross-reactive antibo­
Viral genetic variability (number and variety of viral strains/ dies. Lee et al. found that long-prime, slow-delivery (12 days)
variants that are circulating) also affects vaccine efficacy priming of HIV envelope protein immunogen in rhesus mon­
adversely, as is readily observed with influenza and SARS- keys led to more memory B-cells, higher antibody somatic
CoV2 viruses [147,148]. hypermutation, and more robust immune responses to non-
immunodominant epitopes [154]. Further research is needed
to identify the optimal antigen delivery methods and formula­
4.4. Other factors tions that prolong antigen availability, elicit long-lasting and
Some other ‘environmental factors,’ largely unknown, might protective immunity against influenza and other viruses, and
also affect vaccine-induced immune response. For example, vaccines that provide long-term protection with fewer doses.
there is a drastic difference in immune response to oral Phase 1 of the HIV vaccine clinical trial on this concept is
polio vaccine (OPV) among tropical and temperate coun­ underway (NCT05471076) [124].
tries [144]. One hypothesis that explains this effect is that in Unlike unadjuvanted platforms or those with a single adju­
tropical countries, multiple enteric pathogens are in circula­ vant, replicating viral vectors mimic natural infections and
tion that affect the ‘uptake’ of the vaccine by the immune activate several receptors and inflammatory pathways to sus­
system [149]. tain immune responses. Therefore, early innate signals differ
Even the time of day a vaccine is delivered has been shown by pathogen or platform and may be surrogate endpoints to
to affect immune response robustness. The effect of the circa­ accelerate vaccine development and enable more durable
dian clock on critical immune cell functions, including cytokine protection [124]. Immune correlates of protection by most
production, cell trafficking, dendritic cell activity, and T and modern vaccines are antibodies against pathogen antigens
B cell activity, has been shown; however, the exact mechanism [155]. Since the cellular response is difficult to measure, we
of its effect on the immune response at the cellular or mole­ have mainly focused on humoral antigen response. However,
cular level is still unclear [150,151]. the quantitative analysis and functional characterization of
CD4+ and CD8+ T-cell memory have advanced in the recent
2–3 decades. Multiple methods, such as MHC Class I tetramers,
5. How can we improve the durability of peptide-induced intracellular cytokine labeling, and IFNγ
vaccine-induced protection? ELISPOT, can evaluate the frequency of antigen-specific T-cell
responses [156]. With advanced flow cytometry equipment
5.1. Challenges and the way ahead
and reagents, many T-cell functions and phenotypic markers
Despite the vaccines’ 225-year success in controlling infectious can be evaluated simultaneously. These investigations and
disease, improving quality of life, increasing life expectancy, advanced microarray analysis of T-cell gene expression profiles
and generating substantial economic benefits, it is ironic that have provided information on memory T-cell response [126].
we are still learning how the best vaccines work and how to For instance, antiviral T-cell memory lasts decades after small­
improve vaccine design to improve protective efficacy. Most of pox immunization [157,158] and SARS-CoV-1 infection [159].
the current vaccines have been empirically developed. Vaccine Despite all the information and the growing consensus that
durability assessment is also a ‘wait-and-see’ method due to for the most challenging infectious diseases (e.g. AIDS,
ignorance of immunological memory mechanisms and chal­ malaria, and tuberculosis), a combination of effective patho­
lenges in measuring cellular components that induce long- gen-specific humoral and cell-mediated immunity may be
lasting immune protection [152]. Recent advancements are needed to create an effective vaccine, we are still far from
beginning to reduce our knowledge gaps in underlying harnessing cellular immunity. Identification and validation of
mechanisms of vaccine-mediated protection and long-term T cell-specific correlates of immunity (or co-correlates of
immunity. We may induce more effective and long-lasting immunity of T cells and antibodies needed for vaccine-
immunity with fewer dosages by exploiting these nuances mediated protection) will go a long way in achieving tremen­
and optimizing immune response linkages. dous success in this field.
Natural infection-induced immunity is usually more persis­ New adjuvants should also be explored to prolong vaccine-
tent than vaccine-induced protection. Exploring this disparity induced protection. Inactivated and recombinant vaccines use
is one approach to developing more durable protection vac­ adjuvants to boost vaccine-mediated immune responses,
cines. Beukema et al. [153] prolonged antigen delivery to increasing seroconversion rates, antigen dose sparing, and
replicate natural infection antigen dynamics to improve influ­ fewer doses for similar protection. Alum salts, the most pop­
enza vaccination durability in mice. They compared the ular adjuvants, face the same issues as vaccines: their mechan­
immune response to standard prime-boost immunization ism of action is still debated [160]. Newer adjuvant systems
with a 28-day interval to that of daily doses of whole- may target innate immune system signaling pathways like
inactivated influenza virus vaccine for 14, 21, or 28 days. The Toll-like receptors (TLR) to direct vaccination-induced immu­
28 days of extended antigen administration achieved the high­ nity and better match pathogen-specific immunity require­
est cellular and humoral immune response levels, followed by ments (e.g. skewing of cellular vs. humoral immunity),
21 and 14 days of delivery and prime-boost immunization resulting in a longer-lasting immune response. For example,
EXPERT REVIEW OF VACCINES 403

AS04, a combination of MPL (monophosphoryl lipid A) (a TLR- and their interaction with the host cell, helped experts to
4 agonist) and aluminum hydroxide, is used in an HPV vaccine, design more effective and potent formulations, as shown in
TCV™. This HPV vaccine Cervarix®, when compared to the successful development of RSV vaccines. Some progress in
Cervarix®, on the same vaccine platform (Virus-like Particle, ensuring vaccine-induced immunity’s longevity is also there,
VLP) but with only an aluminum salt (aluminum hydroxy but it is much slower. As discussed in our review, HPV vaccines
phosphate sulfate) as adjuvant, induced a 2- to 9-fold greater shine out in this aspect. Based on the VLP vaccine platform,
antibody response but had a similar serum-antibody decay they have supramolecular nanoparticles with highly repetitive
showing that ASO4 does not substantially change humoral antigen sequences on their surfaces that significantly promote
immune response kinetics [161]. Thus, a better adjuvant sys­ the early B-cell activation and the persistence of GC responses,
tem can help us achieve a ‘tailor-made’ immune response. leading to enhanced long-lasting plasma cell differentiation
Given the theoretical possibility of immune-mediated disease and durable immune response. Thus, despite being inacti­
induction with such an adjuvant, caution is warranted toward vated vaccines, they offer long-term protection, similar to
safety when pursuing this approach. live attenuated vaccines, without needing boosters. Many
other strategies are currently being hotly researched to
improve the durability of vaccine protection. Tweaking con­
6. Conclusion
ventional immunization techniques such as long-prime, slow-
Vaccination research and development have made great delivery (12 days) immunization approach is one such strategy
strides in the last two centuries, yet most vaccines have in this direction, as demonstrated in some animal studies with
been developed empirically. We must still understand their experimental HIV vaccine [154] and influenza vaccine devel­
mechanisms, especially the complicated interaction of opment [153].
innate, humoral, and cellular components and their subcom­ Most currently licensed vaccines have been developed
ponents [162]. Discoveries in this field are beginning to empirically with a ‘develop and then see/assess’ approach.
reduce our knowledge gaps. Similarly, bioengineering tech­ The need of the hour is to develop vaccines strategically.
nologies are evolving rapidly. With nanoparticle and virus- Undoubtedly, there are challenges regarding our understand­
like particle vaccinations, antigen valence and density are ing of immunology and the exact correlates of long-term
finely regulated. Antigen delivery can be controlled and protection. Thus, there is a need to identify ‘protective thresh­
sustained via newer biomaterials. New adjuvants, especially olds’ for different vaccine formulations and bio-markers of
mucosal ones, can activate specific innate immune path­ protective immunity so that minimum antibody levels should
ways. As the mechanisms of immune response durability be maintained above the threshold to avoid a breakthrough
become clearer, we can construct vaccines strategically to infection [107].
provide durable vaccine-induced protection with fewer As elaborated in the review, the two vital immunological
doses. mechanisms that enhance the persistence of immune
responses are memory B cell generations in the germinal
centers and the survival of long-lived plasma cells in the
7. Expert opinion
bone marrow niches. There is a need to recognize factors
Vaccines have contributed immensely to the reduction of that lead to the genesis and persistence of germinal centers
morbidity and mortality associated with infectious diseases, apart from the nature and kinetics of their delivery.
and ultimately reduction in infant/under-five mortality rates, Similarly, identifying early innate signals that differ by
Disability Life Years (DALY), school and work absenteeism, and agents and vaccine platforms could help develop and
thus rise in productivity, the standard of living and life expec­ ensure the survival of long-lasting plasma cells in the
tancy, We have achieved a lot despite having only a handful of bone marrow, providing a lasting immune response.
vaccines that provide long term (>20 years/lifelong) protection Working on adjuvanted SARS-CoV-2 vaccine on non-
with one or two doses; vast majority of vaccines provide human primates, Prabakaran et al. have demonstrated
protection for 7–10 years, and often require multiple doses. a sensitive method to identify and isolate antigen-specific
Quite a few vaccines provide only short-term protection (<5 long-lived plasma cells (LLPCs). Their published work has
years). Enhancing longevity to the vaccine-induced protection shown that spike protein vaccination can elicit and maintain
would undoubtedly act as a ‘force multiplier’ in achieving all LLPCs [163].
the benefits enumerated above. Further, by helping drastically Targeting specific epitopes or genes in antigenic sites
reduce the cost of immunization programs, it has tremendous conserved in different variants can also ensure the durabil­
direct and indirect economic benefits to the nations, adding to ity of protection. Further, there is a need to focus beyond
the reputation of vaccines in particular and medical science in the measurement of antibody production and target key
general. mucosal sites to demonstrate tissue-generated memory
The last few years have seen some rapid strides in the cells at different mucosal tissues, which may help prevent
development of vaccines. The gestation period of developing breakthrough infections and ensure longevity.
a new vaccine has been cut down significantly – from the Identifying solutions to these gaps in our understanding
approval of a concept to the licensing of a product. Thanks should form the future research agenda in the vaccinology
mainly to the advent of new vaccine platforms, like nucleic field. We are confident that we are moving slowly but surely
acid-based mRNA and DNA vaccines, coupled with the from an era of empirically developed vaccines to that of
advanced knowledge of biologically active forms of antigens ‘designer vaccines,’ with particular emphasis on the durability
404 V. M. VASHISHTHA AND P. KUMAR

of protection. In the coming years, the vaccine industry shall flavivirus infection. J Med Virol. 1998;56(2):159–67. doi: 10.1002/
be able to resolve the current shortcomings with the help of (SICI)1096-9071(199810)56:2<159:AID-JMV10>3.0.CO;2-B
advancements in biomaterials and bio-engineering for vaccine 8. Groot H, Riberiro RB. Neutralizing and haemagglutination-inhibiting
antibodies to yellow fever 17 years after vaccination with 17D vaccine.
development. Bull World Health Organ. 1962;27(6):699–707.
9. Niedrig M, Lademann M, Emmerich P, et al. Assessment of IgG
antibodies against yellow fever virus after vaccination with 17D
Funding by different assays: neutralization test, haemagglutination inhibi­
This paper was not funded. tion test, immunofluorescence assay and ELISA. Trop Med
Int Health. 1999 Dec;4(12):867–71.
10. Coulange Bodilis H, Benabdelmoumen G, Gergely A, et al. Persistance
Declaration of interests à long terme des anticorps neutralisants de la fièvre jaune chez les
personnes âgées de 60 ans et plus [Long term persistence of yellow
The authors have no relevant affiliations or financial involvement with fever neutralising antibodies in elderly persons]. Bull Soc Pathol Exot.
any organization or entity with a financial interest in or financial 2011;104(4):260–265. doi: 10.1007/s13149-011-0135-7
conflict with the subject matter or materials discussed in the manu­ 11. Marchesani R, Thomas N, Monath, et al. Comparative safety and
script. This includes employment, consultancies, honoraria, stock own­ immunogenicity of two yellow fever 17D vaccines (ARILVAX and
ership or options, expert testimony, grants or patents received or YF-VAX) in a phase III multicenter, double-blind clinical trial. Am
pending, or royalties. J Trop Med Hyg. 2002;66(5):533–541. doi: 10.4269/ajtmh.2002.66.533
12. World Health Organization. WHO position paper on hepatitis
a vaccines – October 2022. Wkly Epidemiol Rec. 2022;97:493–512.
Reviewer disclosures 13. Espul C, Benedetti L, Cuello H, et al. Persistence of immunity from
Peer reviewers on this manuscript have no relevant financial or other 1 year of age after one or two doses of hepatitis a vaccine given to
relationships to disclose. children in Argentina. Hepat Med. 2012;4:53–60. doi: 10.2147/
HMER.S33847
14. Urueña A, Badano MN, Baré P, et al. Humoral and cellular immune
Author contributions memory response 12 years following single dose vaccination
against hepatitis a in Argentinian children. Vaccine. 2022;40
VMV and PK have substantially contributed to the conception and (1):114–121. doi: 10.1016/j.vaccine.2021.11.037
design of the review article and interpreting the relevant literature, 15. Bianchi FP, Larocca AMV, Bozzi A, et al. Long-term persistence of
and been involved in writing the review article or revised it for intel­ poliovirus neutralizing antibodies in the era of polio elimination:
lectual content. An Italian retrospective cohort study. Vaccine. 2021;39
(22):2989–2994. doi: 10.1016/j.vaccine.2021.04.005
16. World Health Organization. Polio vaccines: WHO position paper.
ORCID Weekly Epidemiol Rec. 2022;97:277–300.
17. Larocca AMV, Bianchi FP, Bozzi A, et al. Long-term immunogenicity
Vipin M. Vashishtha http://orcid.org/0000-0003-1097-117X
of inactivated and oral polio vaccines: an Italian retrospective
cohort study. Vaccines (Basel). 2022 17;10(8):1329. doi: 10.3390/
vaccines10081329
References
18. Bhave S, Sapru A, Bavdekar A, et al. Long term immunogenicity of
Papers of special note have been highlighted as either of interest (•) single dose of live attenuated hepatitis a vaccine in Indian children
or of considerable interest (••) to readers. - results of 15-year follow-up. Indian Pediatr. 2021;58(8):749–752.
1. Morens DM, Taubenberger JK, Fauci AS. Rethinking next-generation doi: 10.1007/s13312-021-2285-4
vaccines for coronaviruses, influenza viruses, and other respiratory 19. Chen Y, Zhou CL, Zhang XJ, et al. Immune memory at 17-years of
viruses. Cell Host Microbe. 2023;31(1):146–157. follow-up of a single dose of live attenuated hepatitis a vaccine.
• This article reviews the factors behind the differences of dur­ Vaccine. 2018;36(1):114–121. doi: 10.1016/j.vaccine.2017.11.036
ability of protection offered by vaccines against the visruses 20. Ott JJ, Irving G, Wiersma ST. Long-term protective effects of hepa­
that cause systemic infection/viremia versus those that multi­ titis a vaccines. A systematic review. Vaccine. 2012;31(1):3–11. doi:
ply in mucosal surfaces only and don’t cause viremia. 10.1016/j.vaccine.2012.04.104
2. Markowitz LE, Preblud SR, Fine PE, et al. Duration of live measles 21. World Health Organization. Human papillomavirus vaccines: WHO
vaccine-induced immunity. Pediatr Infect Dis J. 1990; 9(2): 101–110. position paper (2022 update). Wkly Epidemiol Rec. 2022;97:645–672.
doi: 10.1097/00006454-199002000-00008 22. Porras C, Tsang SH, Herrero R, et al. Efficacy of the bivalent HPV
3. Amanna IJ, Carlson NE, Slifka MK. Duration of humoral immunity to vaccine against HPV 16/18-associated precancer: long-term
common viral and vaccine antigens. N Engl J Med. 2007;357 follow-up results from the Costa Rica vaccine trial. Lancet Oncol.
(19):1903–1915. 2020;21(12):1643–1652. doi: 10.1016/S1470-2045(20)30524-6
• This longitudinal analysis of antibody titers specific for viral 23. Kjaer SK, Nygård M, Sundström K, et al. Final analysis of a 14-year
antigens and nonreplicating antigens for a period of up to 26 long-term follow-up study of the effectiveness and immunogeni­
years gives deep insight into differences in kinetics of anti­ city of the quadrivalent human papillomavirus vaccine in women
body titres of different types of antigens. from four Nordic countries. E-Clin Med. 2020;23:100401. doi: 10.
4. World Health Organization. Rubella vaccines: WHO position paper. 1016/j.eclinm.2020.100401
Wkly Epidemiol Rec. 2020;95:301–324. 24. Olsson SE, Restrepo JA, Reina JC, et al. Long-term immunogenicity,
5. Crooke SN, Riggenbach MM, Ovsyannikova IG, et al. Durability of effectiveness, and safety of nine-valent human papillomavirus vac­
humoral immune responses to rubella following MMR vaccination. cine in girls and boys 9 to 15 years of age: interim analysis after 8
Vaccine. 2020;38(51):8185–8193. doi: 10.1016/j.vaccine.2020.10.076 years of follow-up. Papillomavirus Res. 2020;10:100203. doi: 10.
6. Poland JD, Calisher CH, Monath TP, et al. Persistence of neutralizing 1016/j.pvr.2020.100203
antibody 30-35 years after immunization with 17D yellow fever 25. Kjaer SK, Nygård M, Sundström K, et al. Long-term effectiveness of
vaccine. Bull World Health Organ. 1981;59(6):895–900. the nine-valent human papillomavirus vaccine in Scandinavian
7. Reinhardt B, Jaspert R, Niedrig M, et al. Development of viremia women: interim analysis after 8 years of follow-up. Hum Vaccin
and humoral and cellular parameters of immune activation after Immunother. 2021;17(4):943–949. doi: 10.1080/21645515.2020.
vaccination with yellow fever virus strain 17D: a model of human 1839292
EXPERT REVIEW OF VACCINES 405

26. Shilling H, Garland SM, Atchison S, et al. Human papillomavirus pre­ 46. Madhi SA, Klugman KP, Kuwanda L, et al. Quantitative and quali­
valence and risk factors among Australian women 9–12 years after tative anamnestic immune responses to pneumococcal conjugate
vaccine program introduction. Vaccine. 2021;39(34):4856–4863. doi: vaccine in HIV-infected and HIV-uninfected children 5 years after
10.1016/j.vaccine.2021.07.005 vaccination. J Infect Dis. 2009;199(8):1168–76. doi: 10.1086/597388
27. Aronson NE, Santosham M, Comstock GW, et al. Long-term efficacy 47. World Health Organization. Rabies vaccines: WHO position paper –
of BCG vaccine in American Indians and Alaska Natives: a 60-year April 2018. Wkly Epidemiol Rec. 2018; 93:201–220. doi: 10.1016/j.
follow-up study. JAMA. 2004 5;291(17):2086–91. doi: 10.1001/jama. vaccine.2018.06.061
291.17.2086 48. Briggs DJ, Schwenke JR. Longevity of rabies antibody titre in reci­
28. Nguipdop-Djomo P, Heldal E, Rodrigues LC, et al. Duration of BCG pients of human diploid cell rabies vaccine. Vaccine. 1992;10
protection against tuberculosis and change in effectiveness with (2):125–9. doi: 10.1016/0264-410X(92)90029-J
time since vaccination in Norway: a retrospective population-based 49. Thraenhart O, Kreuzfelder E, Hillebrandt M, et al. Long-term
cohort study. Lancet Infect Dis. 2016;16(2):219–26. doi: 10.1016/ humoral and cellular immunity after vaccination with cell culture
S1473-3099(15)00400-4 rabies vaccines in man. Clin Immunol Immunopathol. 1994;71
29. Mangtani P, Nguipdop-Djomo P, Keogh RH, et al. The duration of (3):287–92. doi: 10.1006/clin.1994.1088
protection of school-aged BCG vaccination in England: a 50. Mansfield KL, Andrews N, Goharriz H, et al. Rabies pre-exposure
population-based case-control study. Int J Epidemiol. 2018;47 prophylaxis elicits long-lasting immunity in humans. Vaccine.
(1):193–201. doi: 10.1093/ije/dyx141 2016;34(48):5959–5967. doi: 10.1016/j.vaccine.2016.09.058
30. Abubakar I, Pimpin L, Ariti C, et al. Systematic review and meta- 51. Chlibek R, Pauksens K, Rombo L, et al. Long-term immunogenicity
analysis of the current evidence on the duration of protection by and safety of an investigational herpes zoster subunit vaccine in
bacillus Calmette-Guérin vaccination against tuberculosis. Health older adults. Vaccine. 2016 3;34(6):863–8. doi: 10.1016/j.vaccine.
Technol Assess. 2013;17(37):1–372, v–vi. 10.3310/hta17370 2015.09.073
31. World Health Organization. Diphtheria vaccine: WHO position 52. Boutry C, Hastie A, Diez-Domingo J, et al. The adjuvanted recom­
paper. Weekly Epidemiol Rec. 2006;91:21–32. binant zoster vaccine confers long-term protection against herpes
32. Bruce MG, Bruden D, Hurlburt D, et al. Antibody levels and protec­ zoster: interim results of an extension study of the pivotal phase 3
tion after hepatitis B vaccine: results of a 30-year follow-up study clinical trials ZOE-50 and ZOE-70. Clin Infect Dis. 2022;74
and response to a booster dose. J Infect Dis. 2016;214(1):16–22. doi: (8):1459–1467. doi: 10.1093/cid/ciab629
10.1093/infdis/jiv748 53. Wendelboe AM, Van Rie A, Salmaso S, et al. Duration of immunity
33. World Health Organization. WHO Immunologic Basis For against pertussis after natural infection or vaccination. Pediatr
Immunization: Module 3: Tetanus. 2018 [accessed 2024 Mar 1]. Infect Dis J. 2005;24(5 Suppl):S58–61. doi: 10.1097/01.inf.
Available from: https://www.who.int/publications/i/item/ 0000160914.59160.41
9789241513616 54. Vashishtha VM, Bansal CP, Gupta SG. Pertussis vaccines: position
34. World Health Organization. Tetanus vaccines: WHO position paper. paper of Indian Academy of Pediatrics (IAP). Indian Pediatr. 2013;50
Weekly Epidemiol Rec. 2017;92:53–76. (11):1001–9. doi: 10.1007/s13312-013-0274-y
35. Ferlito C, Biselli R, Mariotti S, et al. R. Tetanus-diphtheria vaccination 55. Jenkinson D Duration of effectiveness of pertussis vaccine: evi­
in adults: the long-term persistence of antibodies is not dependent dence from a 10-year community study. Br Med J (Clin Res Ed).
on polyclonal B-cell activation and the defective response to 1988;296(6622):612–614. 10.1136/bmj.296.6622.612
diphtheria toxoid re-vaccination is associated to HLADRB1*01. 56. Campbell H, Amirthalingam G, Andrews N, et al. Accelerating con­
Vaccine. 2018;36(45):6718–6725. doi: 10.1016/j.vaccine.2018.09.041 trol of pertussis in England and Wales. Emerg Infect Dis. 2012;18
36. World Health Organization. Varicella and herpes zoster vaccines: (1):38–47. doi: 10.3201/eid1801.110784
WHO position paper, June 2014. Wkly Epidemiol Rec. 2014;89 57. Mueller J, Koutangni T, Guiso N, et al. Comparative efficacy/effec­
(25):265–287. tiveness of schedules in infant immunisation against pertussis,
37. Bayer O, Heininger U, Heiligensetzer C, et al. Metaanalysis of vac­ diphtheria and tetanus: systematic review and meta-analysis.
cine effectiveness in varicella outbreaks. Vaccine. 2007;25(37– Part 2: whole-cell pertussis vaccine. 2014 [accessed 2024 Jan 6].
38):6655–60. doi: 10.1016/j.vaccine.2007.07.010 Available from: http://www.who.int/immunization/sage/meetings/
38. World Health Organization. Position paper. Mumps virus vaccines. 2015/april/6_Report_wP_140813.pdf?ua=1
Wkly Epidemiol Rec. 2007;7:51–60 58. World Health Organization. WHO position paper-pneumococcal
39. Vashishtha VM, Yadav S, Dabas A, et al. IAP position paper on vaccines. Duration of Protection and Revaccination. [accessed
burden of mumps in India and vaccination strategies. Indian 2023 Dec 10]. Available from: https://cdn.who.int/media/docs/
Pediatr. 2015;52(6):505–14. doi: 10.1007/s13312-015-0666-2 default-source/immunization/position_paper_documents/pneumo
40. Demicheli V, Rivetti A, Debalini MG, et al. Vaccines for measles, coccus/ppv23_additional_summary_duration_protection_revacci
mumps and rubella in children. Cochrane Database Syst Rev. nation.pdf?sfvrsn=ddf846b_3.
2012;2:CD004407. doi: 10.1002/14651858.CD004407.pub3 59. Butler JC. Pneumococcal polysaccharide vaccine efficacy. An eva­
41. Cohen C, White JM, Savage EJ, et al. Vaccine effectiveness esti­ luation of current recommendations. JAMA. 1993;270
mates, 2004-2005 mumps outbreak, England. Emerg Infect Dis. (15):1826–1831. doi: 10.1001/jama.1993.03510150060030
2007;13(1):12–17. doi: 10.3201/eid1301.060649 60. Sara E, Oliver SE, Moro P, et al. Epidemiology and prevention of
42. LeBaron CW, Forghani B, Beck C, et al. Persistence of mumps vaccine-preventable diseases. Haemophilus influenzae [accessed
antibodies after 2 doses of measles-mumps-rubella vaccine. 2024 Jan 5]. Available from: https://www.cdc.gov/vaccines/pubs/
J Infect Dis. 2009;199(4):552–560. doi: 10.1086/596207 pinkbook/hib.html#:~:text=Hib%20conjugate%20vaccines%20are%
43. Davidkin I, Jokinen S, Broman M, et al. Persistence of measles, 20highly,children%20who%20are%20fully%20vaccinated
mumps, and rubella antibodies in an MMR-vaccinated cohort: a 61. World Health Organization. Haemophilus influenzae type b (hib)
20-year follow-up. J Infect Dis. 2008 1;197(7):950–6. doi: 10.1086/ vaccination position paper – July 2013. Wkly Epidemiol Rec.
528993 2013;88(39):413–426.
44. World Health Organization. Pneumococcal conjugate vaccines in 62. von Gottberg A, Cohen C, Whitelaw A, et al.; Group for Enteric,
infants and children under 5 years of age: WHO position paper – Respiratory, Meningeal Disease Surveillance in South Africa
February 2019. Wkly Epidemiol Rec. 2019;94:85–104. (GERMS-SA). Invasive disease due to Haemophilus influenzae ser­
45. Le Polain De Waroux O, Flasche S, Prieto-Merino D, et al. The otype b ten years after routine vaccination, South Africa,
efficacy and duration of protection of pneumococcal conjugate 2003-2009. Vaccine. 2012;30(3):565–571. doi: 10.1016/j.vaccine.
vaccines against nasopharyngeal carriage: a meta-regression 2011.11.066.
model. Pediatric Infectious Disease Journal. 2015;34(8):858–864. 63. Ladhani S, Heath PT, Ramsay ME, et al. Long-term immunological
doi: 10.1097/INF.0000000000000717 follow-up of children with Haemophilus influenzae serotype
406 V. M. VASHISHTHA AND P. KUMAR

b vaccine failure in the United Kingdom. Clin Infect Dis. 2009;49 81. Sridhar S, Luedtke A, Langevin E, et al. Effect of Dengue Serostatus
(3):372–80. doi: 10.1086/600292 on Dengue vaccine safety and efficacy. N Engl J Med. 2018;379
64. Sharma H, Yadav S, Lalwani S, et al. Antibody persistence of two (4):327–340. doi: 10.1056/NEJMoa1800820
pentavalent DTwP-HB-Hib vaccines to the age of 15-18 months, 82. Rivera L, Biswal S, Sáez-Llorens X, et al. Three-year efficacy and
and response to the booster dose of quadrivalent DTwP-Hib safety of Takeda’s dengue vaccine Candidate (TAK-003). Clin Infect
vaccine. Vaccine. 2013;31(3):444–447. doi: 10.1016/j.vaccine.2012. Dis. 2022;75(1):107–117. doi: 10.1093/cid/ciab864
11.038 83. Tricou V Efficacy and safety of Takeda’s tetravalent dengue vaccine
65. Gunardi H, Rusmil K, Fadlyana E, et al. DTwP-HB-Hib: antibody Candidate (TAK-003) after 4.5 years of follow-up. Presented at 8th
persistence after a primary series, immune response and safety Northern European Conference of Travel Medicine; Jun 2022
after a booster dose in children 18-24 months old. BMC Pediatr. [accessed 2023 Dec 12]. Available from: https://assets-dam.takeda.
2018;18(1):177. doi: 10.1186/s12887-018-1143-6 com/raw/upload/v1668754472/legacy-dotcom/siteassets/system/
66. Hu Z, Rou YL, Chen C, et al. An epidemiological and serological what-we-do/areas-of-focus/vaccines/info/tides-fact-sheet.pdf
study on duration of protection after meningococcal group 84. World Health Organization. Cholera vaccines: WHO position paper–
a polysaccharide (APS) vaccination. In: Poolman JT, editor. August 2017. Wkly Epidemiol Rec. 2017;92(34):477–498.
Gonococci and Meningococci. Dordrech: Springer; 1988. p. 199– 85. Bhattacharya SK, Sur D, Ali M, et al. 5-year efficacy of a bivalent killed
207. whole-cell oral cholera vaccine in Kolkata, India: a cluster-randomised,
67. Reingold AL, Broome CV, Hightower AW, et al. Age-specific differ­ double-blind, placebo-controlled trial. Lancet Infect Dis. 2013;13
ences in duration of clinical protection after vaccination with (12):1050–1056. doi: 10.1016/S1473-3099(13)70273-1
meningococcal polysaccharide A vaccine. Lancet. 86. Tandan JB, Ohrr H, Sohn YM, et al. Single dose of SA 14-14-2 vaccine
1985;2 (8447):114–118. doi: 10.1016/S0140-6736(85)90224-7 provides long-term protection against Japanese encephalitis: a
68. Andrews N, Borrow R, Miller E Validation of serological correlate of case-control study in Nepalese children 5 years after immunization.
protection for meningococcal C conjugate vaccine by using effi­ Vaccine. 2007; 25(27):5041–5045. doi: 10.1016/j.vaccine.2007.04.052
cacy estimates from post-licensure surveillance in England. Clin 87. Khan SA, Choudhury P, Kakati S, et al. Effectiveness of a single dose
Diagn Lab Immunol. 2003;10(5):780–786. doi: 10.1128/CDLI.10.5. of Japanese encephalitis vaccine among adults, Assam, India,
780-786.2003 2012-2018. Vaccine. 2021;39(35):4973–4978. doi: 10.1016/j.vaccine.
69. Borrow R, Andrews N, Goldblatt D, et al. Serological basis for use of 2021.07.041
meningococcal serogroup C conjugate vaccines in the United 88. Preethi L, Alina MS, Chandran L, et al. Duration of seroprotection of
Kingdom: reevaluation of correlates of protection. Infect Immun. the live attenuated SA-14-14-2 Japanese encephalitis vaccine in
2001;69(3):1568–73. doi: 10.1128/IAI.69.3.1568-1573.2001 children in India. J Travel Med. 2023;30(2):taac147. doi: 10.1093/
70. Nolan T, Booy R, Marshall HS, et al. Immunogenicity and safety of jtm/taac147
a quadrivalent meningococcal ACWY-tetanus toxoid conjugate 89. Kadlecek V, Borja-Tabora CF, Eder-Lingelbach S, et al. Antibody
vaccine 6 years after MenC priming as toddlers. Pediatr Infect Dis persistence up to 3 years after primary immunization with inacti­
J. 2019;38(6):643–650. doi: 10.1097/INF.0000000000002334 vated Japanese encephalitis vaccine IXIARO in Philippine children
71. Cohn AC, MacNeil JR, Harrison LH, et al. Active bacterial core and effect of a booster dose. Pediatr Infect Dis J. 2018;37(9):e233–
surveillance (ABCs) team and MeningNet surveillance partners. e240. doi: 10.1097/INF.0000000000002124
Effectiveness and duration of protection of one dose of 90. Taucher C, Kollaritsch H, Dubischar KL. Persistence of the immune
a meningococcal conjugate vaccine. Pediatrics. 2017;139(2): response after vaccination with the Japanese encephalitis vaccine,
e20162193. doi: 10.1542/peds.2016-2193 ®
IXIARO in healthy adults: a five year follow-up study. Vaccine.
72. Schwartz KL, Kwong JC, Deeks SL, et al. Effectiveness of pertussis 2019;37(19):2529–2531. doi: 10.1016/j.vaccine.2019.03.030
vaccination and duration of immunity. CMAJ. 2016;188(16):E399– 91. Paulke-Korinek M, Kollaritsch H, Kundi M, et al. Persistence of
E406. doi: 10.1503/cmaj.160193 antibodies six years after booster vaccination with inactivated
73. Mills KH Immunity to Bordetella pertussis. Microbes Infect. 2001;3 vaccine against Japanese encephalitis. Vaccine. 2015;33
(8):655–677. doi: 10.1016/S1286-4579(01)01421-6 (30):3600–4. doi: 10.1016/j.vaccine.2015.05.037
74. Klein NP, Bartlett J, Rowhani-Rahbar A, et al. Waning protection 92. World Health Organization. Typhoid vaccines: WHO position
after fifth dose of acellular pertussis vaccine in children. N Engl paper – March 2018. Wkly Epidemiol Rec. 2018;93:153–172.
J Med. 2012;367(11):1012–1019. doi: 10.1056/NEJMoa1200850 93. Klugman KP, Koornhof HJ, Robbins JB, et al. Immunogenicity, efficacy
75. Witt MA, Katz PH, Witt DJ. Unexpectedly limited durability of and serological correlate of protection of salmonella typhi vi capsular
immunity following acellular pertussis vaccination in preadoles­ polysaccharide vaccine three years after immunization. Vaccine.
cents in a North American outbreak. Clin Infect Dis. 2012;54 1996;14(5):435–8. doi: 10.1016/0264-410X(95)00186-5
(12):1730–5. doi: 10.1093/cid/cis287 94. Lin FY, Ho VA, Khiem HB, et al. The efficacy of a Salmonella typhi Vi
76. Bell CA, Russell ML, Drews SJ, et al. Acellular pertussis vaccine conjugate vaccine in two-to-five-year-old children. N Engl J Med.
effectiveness and waning immunity in Alberta, Canada: 2001;344(17):1263–1269.10.1056/NEJM200104263441701
2010-2015, a Canadian Immunization Research Network (CIRN) 95. Voysey M, Pollard AJ Seroefficacy of Vi polysaccharide-tetanus
study. Vaccine. 2019;37(30):4140–4146. doi: 10.1016/j.vaccine. toxoid typhoid conjugate vaccine (Typbar TCV). Clin Infect Dis.
2019.05.067 2018;67(1):18–24. doi: 10.1093/cid/cix1145
77. Klein NP, Bartlett J, Fireman B, et al. Waning Tdap Effectiveness in 96. Vadrevu KM, Raju D, Rani S, et al. Persisting antibody responses to
Adolescents. Pediatrics. 2016;137(3):e20153326. doi: 10.1542/peds. ®
Vi polysaccharide-tetanus toxoid conjugate (Typbar TCV ) vaccine
2015-3326 up to 7 years following primary vaccination of children < 2 years of
78. Koepke R, Eickhoff JC, Ayele RA, et al. Estimating the effectiveness age with, or without, a booster vaccination. Vaccine. 2021 Oct
of tetanus-diphtheria-acellular pertussis vaccine (Tdap) for prevent­ 29;39(45):6682–6690. doi: 10.1016/j.vaccine.2021.07.073
ing pertussis: evidence of rapidly waning immunity and difference 97. Klein NP, Bartlett J, Fireman B, et al. Effectiveness of the live zoster
in effectiveness by Tdap brand. J Infect Dis. 2014;210(6):942–53. vaccine during the 10 years following vaccination: real world
doi: 10.1093/infdis/jiu322 cohort study using electronic health records. BMJ. 2023;383:
79. Acosta AM, DeBolt C, Tasslimi A, et al. Tdap vaccine effectiveness in e076321. doi: 10.1136/bmj-2023-076321
adolescents during the 2012 Washington State pertussis epidemic. 98. World Health Organization. Rotavirus vaccines: WHO position
Pediatrics. 2015;135(6):981–9. doi: 10.1542/peds.2014-3358 paper - July 2021. Weekly Epidemiological Rec, 2021;96:301–319.
80. Briere EC, Pondo T, Schmidt M, et al. Assessment of Tdap vaccina­ 99. Clark A, van Zandvoort K, Flasche S, et al. Efficacy of live oral
tion effectiveness in adolescents in integrated health-care systems. rotavirus vaccines by duration of follow-up: a meta-regression of
J Adolesc Health. 2018;62(6):661–666. doi: 10.1016/j.jadohealth. randomised controlled trials. Lancet Infect Dis. 2019;19(7):717–727.
2017.12.011 doi: 10.1016/S1473-3099(19)30126-4
EXPERT REVIEW OF VACCINES 407

100. Vesikari T, Karvonen A, Ferrante SA, et al. Efficacy of the pentava­ • This chapter describes the immune response to different vac­
®
lent rotavirus vaccine, RotaTeq , in Finnish infants up to 3 years of cines and factors affecting durability of immune response.
age: the Finnish extension study. Eur J Pediatr. 2010;169 118. Chaplin DD. Overview of the immune response. J Allergy Clin
(11):1379–86. doi: 10.1007/s00431-010-1242-3 Immunol. 2010;125(2 Suppl 2):S3–23. doi: 10.1016/j.jaci.2009.12.980
101. Phua KB, Lim FS, Lau YL, et al. Rotavirus vaccine RIX4414 efficacy 119. Wu X, Wu P, Shen Y, et al. CD8+ resident memory T cells and viral
sustained during the third year of life: a randomized clinical trial in infection. Front Immunol. 2018;9:2093. doi: 10.3389/fimmu.2018.
an Asian population. Vaccine. 2012;30(30):4552–7. doi: 10.1016/j. 02093
vaccine.2012.03.030 120. Amanna IJ, Slifka MK, Crotty S Immunity and immunological mem­
102. Vashishtha VM, Kalra A, Choudhury P. Influenza vaccination in ory following smallpox vaccination. Immunol Rev.
India: position paper of Indian Academy of Pediatrics, 2013. 2006;211 1:320–337. doi: 10.1111/j.0105-2896.2006.00392.x
Indian Pediatr. 2013;50(9):867–74. doi: 10.1007/s13312-013- 121. Amanna IJ, Slifka MK. Mechanisms that determine plasma cell life­
0230-x span and the duration of humoral immunity. Immunol Rev.
103. Saha S, Chadha M, Shu Y, et al. Divergent seasonal patterns of 2010;236(1):125–138. doi: 10.1111/j.1600-065X.2010.00912.x
influenza types a and B across latitude gradient in tropical asia. 122. Kato Y, Abbott RK, Freeman BL, et al. Multifaceted effects of antigen
Influenza Other Respir Viruses. 2016;10(3):176–184. doi: 10.1111/irv. Valency on B cell response composition and differentiation in vivo.
12372 Immunity. 2020;53(3):548–563.e8. doi: 10.1016/j.immuni.2020.08.001
104. Young B, Sadarangani S, Jiang L, et al. Duration of Influenza vaccine 123. Heinz FX, Stiasny K. Distinguishing features of current COVID-19
effectiveness: a systematic review, meta-analysis, and vaccines: knowns and unknowns of antigen presentation and
meta-regression of test-negative design case-control studies. modes of action. NPJ Vaccin. 2021;6(1):104. doi: 10.1038/s41541-
J Infect Dis. 2018;217(5):731–741. doi: 10.1093/infdis/jix632 021-00369-6
105. Ferdinands JM, Fry AM, Reynolds S, et al. Intraseason waning of 124. Palin AC, Alter G, Crotty S, et al. The persistence of memory:
influenza vaccine protection: evidence from the US influenza vac­ defining, engineering, and measuring vaccine durability. Nat
cine effectiveness network, 2011-12 through 2014-15. Clin Infect Immunol. 2022;23(12):1665–1668. doi: 10.1038/s41590-022-
Dis. 2016;64(5):544–550. doi: 10.1093/cid/ciw816 01359-z.
106. Regan AK, Fielding JE, Chilver MB, et al. Intra-season decline in • Going beyond the simplistic approach of measuring the neu­
influenza vaccine effectiveness during the 2016 southern hemi­ tralizing antibody titers, the complete landscape of immune
sphere influenza season: a test-negative design study and phylo­ response is described. The current knowledge, knowledge
genetic assessment. Vaccine. 2019;37(19):2634–2641. doi: 10.1016/j. gaps, and opportunities for research on vaccine durability
vaccine.2019.02.027 are detailed in this article.
107. Rambhia KJ, Rambhia MT. Early bird gets the flu: what should be 125. Kumar P. “Mix and match” of COVID vaccines (heterologous boos­
done about waning intraseasonal immunity against seasonal ters) In: Vashishtha V, Kumar P, and Wadhwa A, editors. COVID-19
influenza? Clin Infect Dis. 2019;68(7):1235–1240. doi: 10.1093/cid/ vaccines a comprehensive review. 1st ed. Mumbai: Tree Life Media
ciy748 Publishers, 2022: p. 269–279.
108. Feikin DR, Higdon MM, Abu-Raddad LJ, et al. Duration of effec­ 126. Slifka MK, Amanna I. How advances in immunology provide insight
tiveness of vaccines against SARS-CoV-2 infection and COVID-19 into improving vaccine efficacy. Vaccine. 2014;32(25):2948–57. doi:
disease: results of a systematic review and meta-regression. 10.1016/j.vaccine.2014.03.078
Lancet. 2022;399(10328):924–944. doi: 10.1016/S0140-6736(22) 127. Gordon DM, McGovern TW, Krzych U, et al. Safety, immunogenicity,
00152-0 and efficacy of a recombinant produced Plasmodium falciparum
109. Menegale F, Manica M, Zardini A, et al. Evaluation of waning of circumsporozoite protein-hepatitis B surface antigen subunit
SARS-CoV-2 vaccine-induced immunity: a systematic review and vaccine. J Infect Dis. 1995;171(6):1576–1585. doi: 10.1093/infdis/
meta-analysis. JAMA Netw Open. 2023;6(5):e2310650. doi: 10. 171.6.1576
1001/jamanetworkopen.2023.10650 128. Bejon P, White MT, Olotu A, et al. Efficacy of RTS, S malaria vaccines:
110. Xu S, Li J, Wang H, et al. Real-world effectiveness and factors individual-participant pooled analysis of phase 2 data. Lancet Infect
associated with effectiveness of inactivated SARS-CoV-2 vaccines: Dis. 2013;13(4):319–327. doi: 10.1016/S1473-3099(13)70005-7
a systematic review and meta-regression analysis. BMC Med. 129. Graham BS, Gilman MSA, McLellan JS. Structure-Based Vaccine
2023;21(1):160. doi: 10.1186/s12916-023-02861-3 Antigen Design. Annu Rev Med. 2019 Jan 27;70(1):91–104. doi:
111. Pulendran B, Ahmed R. Immunological mechanisms of vaccination. 10.1146/annurev-med-121217-094234
Nat Immunol. 2011;12(6):509–17. doi: 10.1038/ni.2039 130. Bugya Z, Prechl J, Szénási T, et al. Multiple levels of immunological
112. Kamat D, Mathur A. Vaccine immunology. In: Vashishtha V Kalra A memory and their association with vaccination. Vaccines (Basel).
(editors) IAP Textbook of Vaccines. 2nd ed. New Delhi: Jaypee 2021 Feb 19;9(2):174. doi: 10.3390/vaccines9020174
Brothers Medical Publishers; 2020. p. 31–44. 131. Awate S, Babiuk LA, Mutwiri G. Mechanisms of action of adjuvants.
113. Elsner RA, Shlomchik MJ. Germinal Center and Extrafollicular B Cell Front Immunol. 2013;4:114. doi: 10.3389/fimmu.2013.00114
Responses in Vaccination, Immunity, and Autoimmunity. Immunity. 132. Billeskov R, Beikzadeh B, Berzofsky JA. The effect of antigen dose
2020;53(6):1136–1150. doi: 10.1016/j.immuni.2020.11.006 on T cell-targeting vaccine outcome. Hum Vaccin Immunother.
114. Chen Z, Gao X, Yu D. Longevity of vaccine protection: immunolo­ 2019;15(2):407–411. doi: 10.1080/21645515.2018.1527496
gical mechanism, assessment methods, and improving strategy. 133. Styles TM, Gangadhara S, Reddy PBJ, et al. V2 hotspot optimized
View. 2022;3(1):20200103. MVA vaccine expressing stabilized HIV-1 Clade C envelope Gp140
• This review describes the cellular basis of vaccine-induced delays acquisition of heterologous Clade C Tier 2 challenges in
durable humoral immune response. It also provides the evi­ Mamu-A*01 negative Rhesus Macaques. Front Immunol.
dence and knowledge for the duration of protection by current 2022;13:914969. doi: 10.3389/fimmu.2022.914969
vaccines. 134. Jegaskanda S, Mason RD, Andrews SF, et al. Intranasal Live
115. Stebegg M, Kumar SD, Silva-Cayetano A, et al. Regulation of the Influenza Vaccine Priming Elicits Localized B Cell Responses in
germinal center response. Front Immunol. 2018;9:2469. doi: 10. Mediastinal Lymph Nodes. J Virol. 2018;92(9):e01970–17. doi: 10.
3389/fimmu.2018.02469 1128/JVI.01970-17
116. Elgueta R, de Vries VC, Noelle RJ The immortality of humoral 135. Mok DZL, Chan KR. The effects of pre-existing antibodies on
immunity. Immunol Rev. 2010;236(1):139–150. doi: 10.1111/j.1600- live-attenuated viral vaccines. Viruses. 2020;12(5):520. doi: 10.3390/
065X.2010.00924.x v12050520
117. Siegrist C-A. Vaccine immunology. In: Plotkin S, Orenstein W, 136. Ciarambino T, Para O, Giordano M. Immune system and COVID-19 by
Offit P, editors. Vaccins. 6th ed. Philadelphia, PA: Elsevier- sex differences and age. Womens Health. 2021;17:17455065211022262.
Saunders; 2013; p. 14–32. doi: 10.1177/17455065211022262
408 V. M. VASHISHTHA AND P. KUMAR

137. Cheung F, Apps R, Dropulic L, et al. Sex and prior exposure jointly cell antigen processing. Nat Commun. 2022;13(1):7217. doi: 10.
shape innate immune responses to a live herpesvirus vaccine. Elife. 1038/s41467-022-34897-z
2023;12:e80652. doi: 10.7554/eLife.80652 152. Cohen J. How long do vaccines last? The surprising answers may help
138. Cruz-Tapias P, Castiblanco J, Anaya JM, et al. Major histocompatibility protect people longer. Science Magazine [accessed 2023 Dec 23].
complex: antigen processing and presentation. In: Anaya J, Shoenfeld Y Available from: https://www.science.org/content/article/how-long-
Rojas-Villarraga A, editors. Autoimmunity: from bench to bedside [inter­ do-vaccines-last-surprising-answers-may-help-protect-people-longer
net]. Bogota (Colombia): El Rosario University Press; 2013. Chapter 10. 153. Beukema M, Gong S, Al-Jaawni K, et al. Prolonging the delivery of
Available from: https://www.ncbi.nlm.nih.gov/books/NBK459467/ influenza virus vaccine improves the quantity and quality of the
139. van der Klaauw AA, Horner EC, Pereyra-Gerber P, et al. induced immune responses in mice. Front Immunol.
Accelerated waning of the humoral response to COVID-19 vac­ 2023;14:1249902. doi: 10.3389/fimmu.2023.1249902
cines in obesity. Nat Med. 2023;29(5):1146–1154. doi: 10.1038/ 154. Lee JH, Sutton HJ, Cottrell CA, et al. Long-primed germinal centres
s41591-023-02343-2 with enduring affinity maturation and clonal migration. Nature.
140. Jordan A, Carding SR, Hall LJ. The early-life gut microbiome and 2022;609(7929):998–1004. doi: 10.1038/s41586-022-05216-9
vaccine efficacy. Lancet Microbe. 2022;3(10):e787–e794. doi: 10. 155. Plotkin SA. Vaccines: correlates of vaccine-induced immunity. Clin
1016/S2666-5247(22)00185-9 Infect Dis. 2008 1;47(3):401–409. doi: 10.1086/589862.
141. Moroishi Y, Gui J, Nadeau KC, et al. A prospective study of the • This review describes the correlates of vaccine-induced immu­
infant gut microbiome in relation to vaccine response. Pediatr Res. nity: whether the correlates are absolute, relative of surrogate
2023;93(3):725–731. doi: 10.1038/s41390-022-02154-0 in different situations. The role of cell-mediate immunity as
142. Xu X, Wu Y, Kummer AG, et al. Assessing changes in incubation correlate of protection against disease is also described.
period, serial interval, and generation time of SARS-CoV-2 variants 156. Farber DL, Yudanin NA, Restifo NP. Human memory T cells: gen­
of concern: a systematic review and meta-analysis. BMC Med. eration, compartmentalization and homeostasis. Nat Rev Immunol.
2023;21(1):374. doi: 10.1186/s12916-023-03070-8 2014;14(1):24–35. doi: 10.1038/nri3567
143. Varghese T, Kang G, Steele AD. Understanding rotavirus vaccine 157. Hammarlund E, Lewis MW, Hansen SG, et al. Duration of antiviral
efficacy and effectiveness in countries with high child mortality. immunity after smallpox vaccination. Nature Med. 2003;9
Vaccines (Basel). 2022;10(3):346. doi: 10.3390/vaccines10030346 (9):1131–1137. doi: 10.1038/nm917
144. Parker EP, Ramani S, Lopman BA, et al. Causes of impaired oral 158. Crotty S, Felgner P, Davies H, et al. Cutting edge: long-term B cell
vaccine efficacy in developing countries. Future Microbiol. 2018;13 memory in humans after smallpox vaccination. J Immunol.
(1):97–118. doi: 10.2217/fmb-2017-0128 2003;171(10):4969–4973. doi: 10.4049/jimmunol.171.10.4969
145. Reuman PD, Bernstein DI, Keely SP, et al. Influenza-specific ELISA 159. Duan LJ, Cui XM, Zhu KL, et al. SARS-CoV-2 vaccine-induced anti­
IgA and IgG predict severity of influenza disease in subjects pre­ body and T cell response in SARS-CoV-1 survivors. Cell Rep. 2022;40
screened with hemagglutination inhibition. Antiviral Res. 1990;13 (9):111284. doi: 10.1016/j.celrep.2022.111284
(3):103–10. doi: 10.1016/0166-3542(90)90026-4 160. McKee AS, Munks MW, MacLeod MK, et al. Alum induces innate
146. Lavelle EC, Ward RW. Mucosal vaccines — fortifying the frontiers. Nat immune responses through macrophage and mast cell sensors,
Rev Immunol. 2022;22(4):236–250. doi: 10.1038/s41577-021-00583-2 but these sensors are not required for alum to act as an adjuvant
147. Nuwarda RF, Alharbi AA, Kayser V. An overview of influenza viruses for specific immunity. J Immunol. 2009;183(7):4403–4414. 10.
and vaccines. Vaccines (Basel). 2021;9(9):1032. doi: 10.3390/ 4049/jimmunol.0900164
vaccines9091032 161. Einstein MH, Baron M, Levin MJ, et al. Comparative immunogenicity
148. Carabelli AM, Peacock TP, Thorne LG, et al. SARS-CoV-2 variant and safety of human papillomavirus (HPV)-16/18 vaccine and HPV-
biology: immune escape, transmission and fitness. Nat Rev 6/11/16/18 vaccine: follow-up from months 12–24 in a phase III
Microbiol. 2023;21:162–177. doi: 10.1038/s41579-022-00841-7 randomized study of healthy women aged 18–45 years. Hum
149. Patriarca PA, Wright PF, John TJ. Factors affecting the immunogeni­ Vaccin. 2011;7(12):1343–1358. doi: 10.4161/hv.7.12.18281
city of oral poliovirus vaccine in developing countries: review. Rev 162. Pollard AJ, Bijker EM. A guide to vaccinology: from basic principles
Infect Dis. 1991;13(5):926–39. doi: 10.1093/clinids/13.5.926 to new developments. Nat Rev Immunol. 2021;21(2):83–100. doi:
150. Otasowie CO, Tanner R, Ray DW, et al. Chronovaccination: Harnessing 10.1038/s41577-020-00479-7
circadian rhythms to optimize immunisation strategies. Front Immunol. 163. Prabhakaran M, Matassoli F, Leggat D, et al. Adjuvanted SARS-CoV-
2022;13:977525. doi: 10.3389/fimmu.2022.977525 2 spike protein vaccination elicits long-lived plasma cells in nonhu­
151. Cervantes-Silva MP, Carroll RG, Wilk MM, et al. The circadian clock man primates. Sci Transl Med. 2024;16(728):eadd5960. doi: 10.
influences T cell responses to vaccination by regulating dendritic 1126/scitranslmed.add5960

You might also like