You are on page 1of 8

Available online at www.sciencedirect.

com

ScienceDirect

Vaccination in early life: standing up to the challenges


Elodie Mohr and Claire-Anne Siegrist

The challenge for any vaccine design is to elicit protective responses to this first infant dose are weak, requiring
humoral and/or cytotoxic immunity against life threatening the administration of repeat doses at 1 or 2 months
pathogens while remaining innocuous. Neonatal vaccinology intervals and thus delaying the onset of protection.
faces additional challenges linked to intrinsic peculiarities of the Thus, the 2016 US infant immunization schedule
innate and adaptive neonatal immune system. These include recommends 3 primary doses of DTPa-IPV-HBV/Hib
anti-inflammatory rather than pro-inflammatory responses to and PCV at 2-4-6 months. Infant responses are short-
innate signals, preferential Th2 differentiation limiting the lived, requiring a booster already in the 2nd year of life
induction of Th1 and cytotoxic responses, trends to [1]. The same limitations apply to new vaccines: as an
immunoregulatory responses and weak plasma cell and example, the protective efficacy of the novel RTS,S
germinal centre B cell responses. Recent progresses in our malaria vaccine (although adjuvanted with MPL and
understanding of the molecular bases of these physiological QS21) is higher in children than in infants [2].
peculiarities and of the mode of action of novel adjuvants open
new opportunities to design vaccine formulations and To stand up to their challenges and protect against major
immunization strategies better adapted to the early life period. early life viral (influenza, Respiratory Syncytial Virus
(RSV)), bacterial (pertussis, streptococcus, meningococ-
Address
WHO Collaborative Center for Vaccine Immunology, Departments of
cus) or parasite (malaria) pathogens, neonatal vaccines
Pathology-Immunology and Pediatrics, University of Geneva, should safely elicit strongly protective responses after a
Switzerland single dose — and such responses should be sustained —
or easily boosted.
Corresponding author: Siegrist, Claire-Anne
(Claire-Anne.Siegrist@unige.ch)
Thus, the kinetics, the magnitude and the duration of
protection induced by neonatal vaccines should all be
Current Opinion in Immunology 2016, 41:1–8 enhanced.
This review comes from a themed issue on Vaccines
Edited by Rino Rappuoli and Ennio De Gregorio
The neonatal immune system is adapted to the chal-
lenges of leaving abruptly the almost sterile uterine
For a complete overview see the Issue and the Editorial
environment for the external world, where it faces
Available online 19th April 2016 constant antigenic stimulations. These adaptations re-
http://dx.doi.org/10.1016/j.coi.2016.04.004 quire the implementation of immune tolerance to self-
0952-7915/# 2016 Elsevier Ltd. All rights reserved. antigens and vital foreign elements like food and com-
mensal bacteria, whereas pathogens require the rapid
development of potent immune responses to ensure
immediate and long-term survival. How these seeming-
ly colliding processes, involving a delicate balance be-
tween tolerogenic and pro-inflammatory responses, are
Introduction orchestrated towards the establishment of healthy ho-
Despite the development of vaccines against a growing meostasis [3] remains largely unclear. Hence, vaccina-
spectrum of pathogens, neonates and infants pay a heavy tion requires dedicated strategies to overcome neonatal
toll to infectious diseases. This burden is largely attrib- immaturity [4] and immunoregulatory mechanisms
utable to the unavailability and/or inadequate use of [5,6] while avoiding the excessive inflammation that
vaccine formulations circumventing the intrinsic prop- could lead to tissue damage, allergies or autoimmune
erties of the early life immune system. Indeed, few disorders.
vaccines (BCG, oral polio, hepatitis B) may already be
administered at birth. These prime for effective T cell The neonatal immune system is characterized by anti-
(BCG) or B cell (oral polio, hepatitis B) responses but inflammatory rather than pro-inflammatory responses to
fail to elicit significant primary antibody responses. danger signals and antigens, resulting into the prefer-
Postponing immunization to the 2nd month of life ential differentiation of CD4+ helper T cell (Th) to-
enhances immune response capacity, such that infant wards Th2 cells — antagonizing Th1 and cytotoxic
immunization (against tetanus (T), diphtheria (D), per- responses against intracellular pathogens [7], by the
tussis (Pa), polio (IPV), hepatitis B (HBV), Hemophilus propensity to differentiate into immunoregulatory cells
Influenzae b (Hib), pneumococcus (PCV) and rotavirus) is over effector/memory cells [5,8], by limited plasma cell
routinely initiated at 6–8 weeks of age. Antibody (PC) and germinal centre (GC) B cell responses [9] and

www.sciencedirect.com Current Opinion in Immunology 2016, 41:1–8


2 Vaccines

occasionally by the presence of maternal antibodies (IFN) and supporting the induction of IFN-g-secreting
with immunomodulatory properties [10]. These com- Th1 cells [23,24,25]. Thus, C-lectins and/or neonatal
plex features affect responses to both pathogens and pDC represent potential targets to elicit Th1 antiviral
vaccines. Nonetheless, the ongoing deciphering of the protection in human newborns (see below and
molecular mechanisms underlying the peculiar behav- Figure 1c).
iour of neonatal immune cells is unveiling news
windows of opportunities: newly developed adjuvants Intrinsic features of neonatal CD4+ T cells further in-
and immunization strategies have proven able to crease the difficulties to stimulate adequate immune
improve neonatal responses in preclinical or in vitro responses. Although high levels of miR-181a increases
models, and may eventually apply to neonatal human TCR-induced calcium signalling in neonatal CD4+ T
immunization. cells compared to adult, this signal fails to induce AP-1-
dependent cytokines production and result in anergy
Here we discuss recent progresses in the understanding of [26]. Neonatal human and murine CD4+ T cells differ-
the early life immune system and how some limitations entiate preferentially into Th2 effectors, partly through
may be overcome by the use of specific adjuvants and epigenetic control involving hypermethylation of IFN-g
immunization strategies. locus and hypomethylation of the Th2 locus [12]. Other
mechanisms underlying the resistance to IL-12 and
The early life immune system preferential Th2 neonatal differentiation include the
Because of low exposure to foreign antigens in utero, the storage of a non-secreted IL-4 isoform in the cytoplasm
newborn’s adaptive immune system is mainly composed of human neonatal CD4+ T cells [27] and the expression
of naı̈ve lymphocytes [11,12,13]. Few effector memory of the IL4Ra/IL13Ra heteromeric receptor by neonatal
CD4+ T cells that readily produce cytokines have been Th1 cells, inducing their apoptosis upon IL-4 ligation [7].
identified in human cord blood, but whether and how That the neonatal T cell pool is composed of high
they could be recruited into protective responses remains proportions of recent thymic emigrants [11] may con-
to be determined [14]. Naive lymphocytes require sev- tribute to their lower responsiveness and preferential
eral divisions and differentiation steps, that is at least 1–2 differentiation into Th2 and regulatory T cells
weeks, to become protective effectors [15]. Therefore, [8,28,29]. It is also likely that the low responsiveness
the paucity of antigen-experienced cells confers vulnera- of neonatal cells to the TLR and IL-1/inflammasome
bility to fast replicating pathogens and leaves newborns pathways impacts on the intrinsic ability of T cells to
reliant on their sole innate immune system, equally respond to vaccines and pathogens. Indeed, bacterial
immature [16]. components stimulate T cell cytotoxicity through
TLR and IL-1 signalling [30], and TLR engagement
Innate cells express a range of receptors recognizing is necessary for effector T cells to overcome Treg sup-
conserved danger-associated molecules expressed by pression [31].
pathogens or distressed cells. Upon engagement of these
innate receptors, adult dendritic cells (DC), neutrophils Humoral responses offer several layers of protection
and macrophages produce inflammatory cytokines and differing by their efficacy and duration (Figure 2). In
increase their capacity to present antigens to T cells, early life, low antibody titres and the need for repeated
initiating and shaping adaptive responses. In neonates, vaccine doses reflect the limitations of extrafollicular
responses to two major danger pathways, the Toll-like (EF) and GC-derived plasma cell responses. This
receptors (TLR) signalling and the IL-1/inflammasome results from B cell-intrinsic features, including weaker
pathways, are dampened and fail to induce potent pro- BCR-mediated signals, the lower expression of co-stim-
inflammatory responses — including IL-12p70, the mas- ulatory molecules for T cells and the slow maturation of
ter cytokine for Th1 and cytotoxic responses [17,18,19] marginal zone B cells, a subset of B cells recognizing
(Figure 1a,b). Endosomal TLR ligands (such as viral conserved pathogen-derived molecules to mount rapid
single-strand or double-strand RNA and bacterial CpG T-independent responses in extrafollicular foci [9]. But
oligonucleotides) binding TLR-3, 7/8 and 9, respectively, early life humoral responses are mainly controlled by B-
trigger better responses in neonates and may offer a cell extrinsic limitations. Follicular dendritic cells
strategy to enhance Th1 responses (see below and (FDC) only develop slowly after birth, which delays
Figure 1a,b) [20,21,22]. GC formation [32], and bone marrow stromal cells
provide insufficient survival factors (such as A Prolifer-
Likewise, recent studies with human cord blood cells ation Inducing Ligand (APRIL)) to support post-GC
pinpointed that monocyte-derived DC readily respond long-lived PC [33]. This insufficient accessory support
to co-activation of dectin-1 and TLR7/8 by producing also affects the human gut, where low numbers of IgA+
IL-12p70, and that neonatal plasmacytoid DC (pDC) PC correlate with poor expression of APRIL until one
respond to the viral RNA content of trivalent inacti- month of life, when both IgA+ PC and APRIL appear
vated influenza vaccines by producing Type interferons [34]. Most importantly, the expansion of T follicular

Current Opinion in Immunology 2016, 41:1–8 www.sciencedirect.com


Vaccination in early life Mohr and Siegrist 3

Figure 1

(a) Cell death (b) (c)


MF59 Alum Uric acid dsDNA
CpG BCG Curdlan BCG CAF01
Zymosan

TLR1
R-848 TLR2 Dection-1 Mincle
TLR5 FcγR
PuR TLR4 CD14 TLR6 IL-1R

4. Syk Syk
IL-1 ASC/CARD Lipid rafts
cytokine family
3. TLR9
MyD88
TLR7/8
2. TLR3 CARD9
Inflammasome
TRIF 5. MALT1 Bcl10
ATP ENDOSOME
Cathepsin B
LYSOSOME
Pro-IL-1 Pro NOD-like NFκB NFκB
cytokine family IRF3 IRF7
caspase-1 receptors
1. 6.
CYTOSOL CYTOSOL
NUCLEUS NUCLEUS Inflammatory
Pro-IL-1 Pro-caspase-1 IL-12p70 TNFα Type I IFNs IL-12p70 Type I IFNs cytokines

7.
NLR pathway TLR pathway CLR pathway

Current Opinion in Immunology

Innate receptors signalling in neonates and adjuvant targets. Black arrows indicate signalling pathways and blue arrows adjuvant targets. Bold
arrows are confirmed effective pathways in neonates, while dashed arrows and elements represent suboptimal pathways. (a) The IL-1/
inflammasome and (b) the TLR signalling modules are two essential synergic components of the danger-sensing machinery, which trigger the
production of inflammatory cytokines such as IL-1b, TNF-a, Type I IFN (IFNa and IFNb), IL6 and IL-12p70. (a) The IL-1/inflammasome pathways
requires two signals: (1) from the TLR, inducing the expression of pro-forms of IL-1 family members (IL-1b, IL-18, IL-33) and caspase 1 by NFkB
activation, and (2) a second signal mediating the maturation of caspase-1 and inflammasome assembly (composed by a nucleotide-binding
oligomerization domain receptors or NOD-like receptors (NLRs), the adaptor apoptosis-associated speck-like protein containing a CARD domain
(ASC), and caspase-1). This second signal results from the ligation of intracellular NOD-like receptor sensing RNAs or the binding of crystals to
lipid rafts provoking lysosome disruption, and subsequent release of cathepsin B in the cytosol, and/or ATP triggering autocrine activation through
purine receptors (PuR). (3) The active inflammasome releases active IL-1 family members. Alum adjuvants likely act directly by binding to lipid
rafts, and indirectly by inducing cell death and release of natural TLR agonists and uric acid, providing signals 1 and 2 respectively [38]. MF59 is
thought to act through ATP/PuR to initiate activation of innate immune cells at the site of injection [63]. In neonates, the limited induction of pro-
IL-1 and pro-caspase 1 expression [48] and the limited cleavage of pro-IL1b by the inflammasome [18] blunt the IL1/inflammasome responses.
(b) Toll-like receptors (TLR) bind pathogen-derived components at the cell surface (TLR1-2, 4–6) or in the endosomes (TLR3, 7/8, 9) where they
recognize microbial-derived and host-derived nucleic acids. TLR4 can also be endocytosed (not depicted). TLR signalling (4) occurs through the
engagement of the intracellular Toll-IL-1 resistance domain (TIR) with the adaptor proteins MyD88 or TRIF (5). This activates IL-1R-associated
(IRAKs) and TNF-associated (TRAFs) factors (not depicted), leading to the activation of nuclear factor kB (NFkB) and Interferon-regulatory factor
(IRF) 3 and/or 7 (6) and IL-12, TNFa and type I IFN responses (7). Several steps limit neonatal TLR-mediated pro-inflammatory responses: TLR
signalling is antagonized by an inhibitory pathway triggered by high plasma ADP [68] and possibly blunted by lower expression of the TLR4/TLR2
co-receptor CD14 [20] and MyD88 [20]. Less effective IRF-7 nuclear translocation and DNA binding of IRF3 [17] limit IL-12 and type I IFN
responses. Interestingly, TLR7/8 agonists are refractory to ADP inhibition and thus active on neonatal cells [48]. (c) Dectin-1 and Mincle signalling.
Dectin-1 and Mincle are innate surface receptors of the C-lectin family. They bind b-glucans on the cell wall of fungi and bacteria and the cord
factor (TDB) expressed on the surface of M. tuberculosis, respectively. Dectin-1 signals directly through its cytoplasmic tyrosine-based activation
motif (ITAM), recruiting the Syk, while Mincle uses ITAM on the FcRg chain. Syk activates the Card9–Bcl10–MALT1 complex, triggering NF-kB and
subsequent production of pro-inflammatory cytokines. These pathways have proven functional in humans in cord blood monocyte-derived and
pDC stimulated with Curdlan [23], in neonatal and infant mice immunized with the TDB-including CAF01 adjuvant [56,57] and following BCG
immunization [60].

helper (TFH) cells, which are critical for effective GC formulations or adjuvants improving neonatal TFH
responses, was recently identified as a key limiting responses is a major challenge to induce the GC
factor for the development of early life GC responses responses required for the establishment of long-lived,
[35,36,37]. Therefore, the development of vaccine high affinity effectors (Figure 2).

www.sciencedirect.com Current Opinion in Immunology 2016, 41:1–8


4 Vaccines

Figure 2

DURATION OF PROTECTION

Extrafollicular responses Germinal center responses Memory cells

Memory CXCR5
Short lived
plasma cells B cells Long lived
OX40 OX40L
plasma cells
ICOS ICOSL
TCR PP-MHC II
TFH CD40
CD40L
cells IL-21R
IL-21 BCR

Ag 2.
3. FDC 4.
BCR
GC B
Cytokines Receptors cells Memory
GC B B cells
TCR PP-MHC II
GC B GC B
CD40L CD40

cycling
CD4+ T 1.
B cells
helper cells

Current Opinion in Immunology

B cells differentiation towards plasma cell and memory B cells in early life vaccine responses. Dotted arrows/lines represent the pathways/cells
reduced in neonates. In the B cell follicles of the lymph nodes draining the injection site, B cells binding to their cognate antigen (Ag) through their
B Cell Receptor (BCR) migrate to the interface between the B cell and the T cell zones, where they interact with activated CD4 T helper (Th) cells
(1). Th/B cell interactions (involving MHC class II/Ag-derived peptides on B cells and TCRab/CD4 on T cells) deliver B cell survival and activation
signals through CD40/CD40L. Activated B cells proliferate and may differentiate either in situ (extrafollicular (EF) responses) (2) or into germinal
centres (3). In early life, most B cells differentiate along the EF pathway and into quiescent memory B cells (MBC) rather than plasma cells (PC).
This generates a low and transient antibody response of weak affinity for the vaccine Ag. Antigen-specific B cells blasts which receive appropriate
activation signals upregulate the BCL-6 transcription factor and migrate back to the B cell follicles, where they seed the germinal centre (GC)
reaction at the vicinity of follicular DC (FDC) retaining the Ag captured in immune complexes (3). In these structures, GC B cells proliferate and
undergo affinity maturation through somatic hypermutation, selection and proliferation. This process requires close interactions of B cells with T
follicular helper cells (TFH), a subset of T helper cells homing to the GC through CXCR5/CXCL13 chemotaxis and delivering specific helper and
survival signals to GC B cells, and with FDCs. In early life, the paucity of FDCs and suboptimal TFH/GC B cell interactions fail to deliver sufficient
B cell activation/survival signals, restricting the GC reaction. GC-derived B cells may differentiate into MBC or long-lived PC capable of surviving
in specific BM niches. In early life, GC-derived B cells preferentially differentiate into MBCs and away from long-lived PC, the early life BM failing
to provide sufficient PC survival signals. Adjuvants capable of inducing potent GC responses and long-lived BMPCs in early life remain to be
identified.

Immunization strategies for early life release of prostaglandins. A body of evidence in vitro
protection indicates that alum uses components of the inflamma-
Understanding the factors and mechanisms underlying some IL-1/caspase 1 pathways [38], the latest discovered
the specific patterns of early life innate and adaptive being the NEK kinase 7 which promotes inflammasome
responses progressively paves the way towards the iden- assembly [39,40]. Alum is insufficient to promote effec-
tification of novel vaccine and adjuvant formulations tive plasma cell responses or the expansion of Tfh cells
which fail to be identified by studies performed on adults. and GC B cells in mouse neonates [35,37], but it
effectively induces memory B cells, likely along the
Alum, the adjuvant currently used in infant vaccines, EF pathway, and triggers affinity maturation already in
exacerbates the neonatal Th2 bias by promoting the neonates [41]. These limitations fit in well with the

Current Opinion in Immunology 2016, 41:1–8 www.sciencedirect.com


Vaccination in early life Mohr and Siegrist 5

observation that inflammasome IL-1/caspase 1 pathways adults [59]. Together with TLR ligands, TDB is a critical
are dampened in human neonates (Figure 1) [18]. How- adjuvant factor of the Bacillus Calmette–Guérin (BCG)
ever, the exact mechanisms of alum adjuvanticity in vivo vaccine, which shows remarkable ability to promote Th1
are still matter of discussions, may differ between innate and antibody responses when used concomitantly with
and adaptive responses, and could involve alternative other vaccine antigens [60] (Figure 1b,c).
pathways as yet to be discovered and assessed in neonatal
life [38]. Nonetheless, alum may prove an appropriate MF59, a squalene-based emulsion developed by Novartis
vehicle for the retention/sustained delivery of early life Vaccines (now GSK), markedly increased Ab responses
immunomodulators. and protection against influenza in 6-36 months infants
[61]. A recent study using a system biology approach
TLR agonists have been extensively tested as immuno- unravelled this adjuvanticity to be mediated by the
modulators (reviewed in [42]), and early life responsive- induction of more homogenous, robust and more adult-
ness to TLR agonists assessed in vitro and occasionally in like gene signatures [62]. Part of the adjuvanticity of
vivo [43,44]. They may mediate B cell adjuvanticity by MF59 relies on ATP release by muscle cells at the site
acting on multiple innate cells including DC (to promote of injection and the subsequent strong recruitment of
the activation of T helper cells) and B cells, which express CD11b+ innate cells encompassing neutrophils and
subsets of TLRs (TLR2, TLR4, TLR7 and TLR9 in monocytes [63] (Figure 1a). Studies in mice demonstrat-
mouse and TLR2, TLR7 and TLR9 in human), and ed that MF59 mediates its B cell adjuvanticity through
respond to their agonists by activation, proliferation, the induction of potent TFH/GC responses in infants —
cytokine secretion and PC differentiation [45,46]. In but fails to do so in neonates in which additional limita-
human, an aluminium-adsorbed TLR2 ligand-containing tions remain to be overcome [36].
Hib vaccine promoted antibody responses when given at
2 months of age — but not in neonates and to the Few other B cell adjuvants have been tested in early life.
detriment of memory responses [43]. Its development A polysaccharide adjuvant based on delta inulin was
was thus not pursued. MPL (Monophosphoryl Lipid A), a reported to increase mainly IgM Ab to influenza, with
TLR4 ligand, synergizes with aluminium salts to improve no suggestion of an induction of GC responses [64]. The
the magnitude and duration of B cell responses to hepa- nontoxic mutant of E. coli heat-labile enterotoxin LT-
titis B and HPV vaccines in adults [47]. The ontogeny of K63 adjuvant enhanced anti-pneumococcal neonatal
TLR4 responses has not been characterized other than in responses, expanding the FDC network and inducing
vitro [43,48]. However, a combination of MPL and QS21 some GC responses [52]. Its clinical development was
(quillaja saponaria 21) enhanced the immunogenicity of interrupted after the occurrence of facial nerve paralysis
the RTS,S malaria vaccine in infants [2]. Studies in following intranasal immunization, but this provided a
mouse suggest that its efficacy results from enhanced DC proof of concept that some GC responses may be elicited
and monocyte activation [49]. TLR7/8 agonists exert through appropriate early life adjuvantation.
potent adjuvanticity in adult mice [50], have entered
clinical development and have been identified as unique- Conclusion
ly capable of activating human neonatal antigen-present- Past vaccine strategies relied on the empirical use of
ing cells [19,21,51], suggesting promising early life attenuated pathogens and aluminium-based adjuvants,
adjuvanticity. TLR9 agonists have been extensively stud- whose mechanism of action remained obscure for more
ied, mainly in addition to aluminium salts, and shown to than 80 years [38]. Today, the knowledge acquired in
increase early life murine Ab responses [32,35,52,53]. microbiology and immunology allows the identification of
Nucleotide oligomerization domain (Nod)-like-receptor the specific pathways and factors that uniquely shape the
agonists elicit innate neonatal responses in whole-blood early life immune system and novel tools, including
assays, but were not yet tested in vivo [54]. system biology approaches, are being applied to study
these critical issues [62]. This enables hypotheses-driven
C-type lectin agonists, which activate macrophages and approaches towards the identification of vaccine formula-
DC through the Syk-Card9 pathway [55] (Figure 1c), are tions specifically designed to stand up to the challenge of
promising early life adjuvants. On human neonatal DC, eliciting more rapid, effective and sustained vaccine
Mincle agonists (such as trehalose 6,6 dimycolate (TDB)) responses as early in life as required by the risks of
and Dectin-1 agonists (such as zymosan or curdlan) en- exposure. Consequently, a growing number of vaccines
hanced DC activation and Th1 polarization, in synergy and adjuvants are being tested in early life animal models,
with TLR agonists [23]. In vivo, TDB induces potent with human cord blood cells, and eventually in clinical
Th1/Th17 neonatal responses through the exquisite tar- trials including children [65].
geting of a small number of DC when formulated in
cationic liposomes (CAF01) [56,57] which are being de- The road will be long until novel vaccines are demon-
veloped as a versatile vaccine adjuvant platform [58] strated sufficiently effective and safe to be used already in
eliciting prolonged CD4+ T cell responses in human neonates — when required by high risks of exposure.

www.sciencedirect.com Current Opinion in Immunology 2016, 41:1–8


6 Vaccines

Meanwhile, maternal immunization has re-emerged as a 13. Duchamp M, Sterlin D, Diabate A, Uring-Lambert B, Guerin-El
 Khourouj V, Le Mauff B, Monnier D, Malcus C, Labalette M,
safe and powerful strategy to protect against early life Picard C: B-cell subpopulations in children: national reference
death, be it from influenza [66] or pertussis [67], and values. Immun Inflamm Dis 2014, 2:131-140.
This article provides a valuable reference for the relative proportions of
possibly in a near future from RSV or Group B Strepto- naı̈ve and memory B cells to be expected in healthy individuals from early
coccus. age to adulthood.
14. Zhang X, Mozeleski B, Lemoine S, Deriaud E, Lim A, Zhivaki D,
Acknowledgements  Azria E, Le Ray C, Roguet G, Launay O et al.: CD4 T cells with
effector memory phenotype and function develop in the sterile
environment of the fetus. Sci Transl Med 2014, 6:238ra272.
Research work in the Center for Vaccinology is supported by the Swiss
This study assessing human fetuses reveals unexpected populations of
National Research Foundation, the European Union’s Seventh Framework effector memory CD4+ T cells in cord blood, suggesting that their
Programme (EU FP7) projects ADITEC (HEALTH-F4-2011-280873), the appropriate stimulation might accelerated the induction of neonatal
EU Horizon 2020 project TBVAC 2020 (Grant No. 643381); the Innovative responses.
Medicines Initiative (IMI) Joint Undertaking (JU) VSV-EBOVAC and the
Fondation Mérieux. 15. Ademokun AA, Dunn-Walters D: Immune responses: primary
and secondary. Encyclopedia of Life Sciences. Chichester: John
Wiley & Sons, Ltd; 2010.

References and recommended reading 16. Levy O: Innate immunity of the newborn: basic mechanisms
Papers of particular interest, published within the period of review, and clinical correlates. Nat Rev Immunol 2007, 7:379-390.
have been highlighted as: 17. Kollmann TR, Levy O, Montgomery RR, Goriely S: Innate immune
function by Toll-like receptors: distinct responses in
 of special interest newborns and the elderly. Immunity 2012, 37:771-783.
 of outstanding interest
18. Sharma AA, Jen R, Kan B, Sharma A, Marchant E, Tang A,
1. ACIP: Immunization Schedules for Persons Age Birth through  Gadawski I, Senger C, Skoll A, Turvey SE et al.: Impaired NLRP3
18 Years. 2015–2016:. http://www.cdc.gov/vaccines/schedules/ inflammasome activity during fetal development regulates IL-
hcp/child-adolescent.html. 1beta production in human monocytes. Eur J Immunol 2015,
45:238-249.
2. Rts SCTP: Efficacy and safety of the RTS,S/AS01 malaria This study identifies a defect in pro-caspase 1 cleavage that contributes
 vaccine during 18 months after vaccination: a phase to the low responses of neonatal cells to adjuvants targeting the IL-1/
3 randomized, controlled trial in children and young infants at inflammasome pathways.
11 African sites. PLoS Med 2014, 11:e1001685.
This report demonstrates the capacity of a novel combined adjuvant 19. Corbett NP, Blimkie D, Ho KC, Cai B, Sutherland DP, Kallos A,
(liposomes containing MPL and saponin) to raise some protective Crabtree J, Rein-Weston A, Lavoie PM, Turvey SE et al.: Ontogeny
responses in infants and young children. of Toll-like receptor mediated cytokine responses of human
blood mononuclear cells. PLoS ONE 2010, 5:e15041.
3. Ghazal P, Dickinson P, Smith CL: Early life response to infection.
Curr Opin Infect Dis 2013, 26:213-218. 20. Morris MC, Surendran N: Neonatal vaccination: challenges and
intervention strategies. Neonatology 2016, 109:161-169.
4. Dowling DJ, Levy O: Ontogeny of early life immunity. Trends
 Immunol 2014, 35:299-310. 21. Ganapathi L, Van Haren S, Dowling DJ, Bergelson I, Shukla NM,
This outstanding review gives a comprehensive overview of the current Malladi SS, Balakrishna R, Tanji H, Ohto U, Shimizu T et al.: The
understanding of the neonatal immune system. The authors focus parti- imidazoquinoline Toll-like receptor-7/8 agonist hybrid-2
cularly on innate cells and receptors functionality in neonates, which they potently induces cytokine production by human newborn and
contributed substantially to elucidate. adult leukocytes. PLOS ONE 2015, 10:e0134640.
5. Ndure J, Flanagan KL: Targeting regulatory T cells to improve 22. Kim D, Niewiesk S: Synergistic induction of interferon alpha
vaccine immunogenicity in early life. Front Microbiol 2014,  through TLR-3 and TLR-9 agonists stimulates immune
5:477. responses against measles virus in neonatal cotton rats.
Vaccine 2014, 32:265-270.
6. Gervassi AL, Horton H: Is infant immunity actively suppressed This study identifies a new combination of TLR agonists (TLR3 and TLR9)
or immature? Virology (Auckl) 2014:1-9. for neonatal immunization to elicit interferon alpha.
7. Zaghouani H, Hoeman CM, Adkins B: Neonatal immunity: faulty 23. Lemoine S, Jaron B, Tabka S, Ettreiki C, Deriaud E, Zhivaki D, Le
T-helpers and the shortcomings of dendritic cells. Trends  Ray C, Launay O, Majlessi L, Tissieres P et al.: Dectin-1 activation
Immunol 2009, 30:585-591. unlocks IL12A expression and reveals the TH1 potency of
8. Wang G, Miyahara Y, Guo Z, Khattar M, Stepkowski SM, Chen W: neonatal dendritic cells. J Allergy Clin Immunol 2015, 136:1355-
‘‘Default’’ generation of neonatal regulatory T cells. J Immunol 1368 e1315.
2010, 185:71-78. This study identifies a new cellular and molecular pathway functional in
cord blood cells which could be targeted to elicit stronger neonatal Th1
9. Siegrist CA, Aspinall R: B-cell responses to vaccination at the responses.
extremes of age. Nat Rev Immunol 2009, 9:185-194.
24. Zhang X, Casartelli N, Lemoine S, Mozeleski B, Azria E, Le Ray C,
10. Niewiesk S: Maternal antibodies: clinical significance, Schwartz O, Launay O, Leclerc C, Lo-Man R: Plasmacytoid
mechanism of interference with immune responses, and dendritic cells engagement by influenza vaccine as a
possible vaccination strategies. Front Immunol 2014, 5:446. surrogate strategy for driving T-helper type 1 responses in
human neonatal settings. J Infect Dis 2014, 210:424-434.
11. Thome JJ, Bickham KL, Ohmura Y, Kubota M, Matsuoka N,
 Gordon C, Granot T, Griesemer A, Lerner H, Kato T et al.: Early-life 25. Zhang X, Lepelley A, Azria E, Lebon P, Roguet G, Schwartz O,
compartmentalization of human T cell differentiation and Launay O, Leclerc C, Lo-Man R: Neonatal plasmacytoid
regulatory function in mucosal and lymphoid tissues. Nat Med dendritic cells (pDCs) display subset variation but can elicit
2016, 22:72-77. potent anti-viral innate responses. PLoS ONE 2013, 8:e52003.
This study provides a comprehensive view of CD4+ T cell subsets in
neonates and infants compared to young adults. This will be very valuable 26. Palin AC, Ramachandran V, Acharya S, Lewis DB: Human
to take into account the ontogeny of the T cell compartments when  neonatal naive CD4+ T cells have enhanced activation-
designing neonatal vaccine schedules or strategies. dependent signaling regulated by the microRNA miR-181a. J
Immunol 2013, 190:2682-2691.
12. Adkins B: Heterogeneity in the CD4 T cell compartment and the This study highlights a deficit of AP-1 induction under TCR signalling in
variability of neonatal immune responsiveness. Curr Immunol neonates that provides a possible explanation as to why neonatal T cells
Rev 2007, 3:151-159. become anergic upon stimulation.

Current Opinion in Immunology 2016, 41:1–8 www.sciencedirect.com


Vaccination in early life Mohr and Siegrist 7

27. Hebel K, Weinert S, Kuropka B, Knolle J, Kosak B, Jorch G, antibody avidity profiles after early-life immunization with
 Arens C, Krause E, Braun-Dullaeus RC, Brunner-Weinzierl MC: protein vaccines. Eur J Immunol 2002, 32:752-760.
CD4+ T cells from human neonates and infants are poised
spontaneously to run a nonclassical IL-4 program. J Immunol 42. Coffman RL, Sher A, Seder RA: Vaccine adjuvants: putting
2014, 192:5160-5170. innate immunity to work. Immunity 2010, 33:492-503.
This articles reports a new unconventional pathway for explaining the
preferential Th2 polarization of human neonatal T cells, through the 43. Levy O, Goriely S, Kollmann TR: Immune response to vaccine
cytoplasmic storage of preformed IL-4. adjuvants during the first year of life. Vaccine 2013, 31:2500-2505.

28. Berkley AM, Hendricks DW, Simmons KB, Fink PJ: Recent thymic 44. Nguyen M, Leuridan E, Zhang T, De Wit D, Willems F, Van Damme P,
emigrants and mature naive T cells exhibit differential Goldman M, Goriely S: Acquisition of adult-like TLR4 and TLR9
DNA methylation at key cytokine loci. J Immunol 2013, responses during the first year of life. PLoS ONE 2010, 5:e10407.
190:6180-6186. 45. Bekeredjian-Ding I, Jego G: Toll-like receptors—sentries in the
29. Paiva RS, Lino AC, Bergman ML, Caramalho I, Sousa AE, B-cell response. Immunology 2009, 128:311-323.
Zelenay S, Demengeot J: Recent thymic emigrants are the
46. Hou B, Saudan P, Ott G, Wheeler ML, Ji M, Kuzmich L, Lee LM,
preferential precursors of regulatory T cells differentiated in
Coffman RL, Bachmann MF, DeFranco AL: Selective utilization
the periphery. Proc Natl Acad Sci U S A 2013, 110:6494-6499.
of Toll-like receptor and MyD88 signaling in B cells for
30. O’Donnell H, Pham OH, Li LX, Atif SM, Lee SJ, Ravesloot MM, enhancement of the antiviral germinal center response.
Stolfi JL, Nuccio SP, Broz P, Monack DM et al.: Toll-like receptor Immunity 2011, 34:375-384.
and inflammasome signals converge to amplify the innate
47. Didierlaurent AM, Morel S, Lockman L, Giannini SL, Bisteau M,
bactericidal capacity of T helper 1 cells. Immunity 2014,
Carlsen H, Kielland A, Vosters O, Vanderheyde N, Schiavetti F
40:213-224.
et al.: AS04, an aluminum salt- and TLR4 agonist-based
31. Schenten D, Nish SA, Yu S, Yan X, Lee HK, Brodsky I, Pasman L, adjuvant system, induces a transient localized innate immune
Yordy B, Wunderlich FT, Bruning JC et al.: Signaling through the response leading to enhanced adaptive immunity. J Immunol
adaptor molecule MyD88 in CD4+ T cells is required to 2009, 183:6186-6197.
overcome suppression by regulatory T cells. Immunity 2014,
40:78-90. 48. Philbin VJ, Dowling DJ, Gallington LC, Cortes G, Tan Z, Suter EE,
Chi KW, Shuckett A, Stoler-Barak L, Tomai M et al.:
32. Pihlgren M, Tougne C, Bozzotti P, Fulurija A, Duchosal MA, Imidazoquinoline Toll-like receptor 8 agonists activate human
Lambert PH, Siegrist CA: Unresponsiveness to lymphoid- newborn monocytes and dendritic cells through adenosine-
mediated signals at the neonatal follicular dendritic cell refractory and caspase-1-dependent pathways. J Allergy Clin
precursor level contributes to delayed germinal center Immunol 2012, 130:195-204 e199.
induction and limitations of neonatal antibody responses to T-
dependent antigens. J Immunol 2003, 170:2824-2832. 49. Didierlaurent AM, Collignon C, Bourguignon P, Wouters S,
Fierens K, Fochesato M, Dendouga N, Langlet C, Malissen B,
33. Belnoue E, Pihlgren M, McGaha TL, Tougne C, Rochat AF, Lambrecht BN et al.: Enhancement of adaptive immunity by the
Bossen C, Schneider P, Huard B, Lambert PH, Siegrist CA: APRIL human vaccine adjuvant AS01 depends on activated dendritic
is critical for plasmablast survival in the bone marrow and cells. J Immunol 2014, 193:1920-1930.
poorly expressed by early-life bone marrow stromal cells.
Blood 2008, 111:2755-2764. 50. Vasilakos JP, Tomai MA: The use of Toll-like receptor 7/8 agonists
as vaccine adjuvants. Expert Rev Vaccines 2013, 12:809-819.
34. Gustafson CE, Higbee D, Yeckes AR, Wilson CC, De Zoeten EF,
Jedlicka P, Janoff EN: Limited expression of APRIL and its 51. Levy O, Suter EE, Miller RL, Wessels MR: Unique efficacy of Toll-
receptors prior to intestinal IgA plasma cell development like receptor 8 agonists in activating human neonatal antigen-
during human infancy. Mucosal Immunol 2014, 7:467-477. presenting cells. Blood 2006, 108:1284-1290.

35. Mastelic B, Kamath AT, Fontannaz P, Tougne C, Rochat AF, 52. Bjarnarson SP, Adarna BC, Benonisson H, Del Giudice G,
 Belnoue E, Combescure C, Auderset F, Lambert PH, Tacchini- Jonsdottir I: The adjuvant LT-K63 can restore delayed
Cottier F et al.: Environmental and T cell-intrinsic factors limit maturation of follicular dendritic cells and poor persistence of
the expansion of neonatal follicular T helper cells but may both protein- and polysaccharide-specific antibody-secreting
be circumvented by specific adjuvants. J Immunol 2012, cells in neonatal mice. J Immunol 2012, 189:1265-1273.
189:5764-5772.
This key study identified T follicular helper cells as a leverage point that 53. Gracia A, Polewicz M, Halperin SA, Hancock RE, Potter AA,
limit/may improve humoral responses to vaccines in early life. Babiuk LA, Gerdts V: Antibody responses in adult and neonatal
BALB/c mice to immunization with novel Bordetella pertussis
36. Mastelic Gavillet B, Eberhardt CS, Auderset F, Castellino F, vaccine formulations. Vaccine 2011, 29:1595-1604.
 Seubert A, Tregoning JS, Lambert PH, de Gregorio E, Del
Giudice G, Siegrist CA: MF59 mediates its B cell adjuvanticity 54. Lisciandro JG, Prescott SL, Nadal-Sims MG, Devitt CJ, Pomat W,
by promoting T follicular helper cells and thus germinal Siba PM, Tulic MC, Holt PG, Strickland D, van den Biggelaar AH:
center responses in adult and early life. J Immunol 2015, Ontogeny of Toll-like and NOD-like receptor-mediated innate
194:4836-4845. immune responses in Papua New Guinean infants. PLoS ONE
See annotation to Ref. [35]. 2012, 7:e36793.

37. Debock I, Jaworski K, Chadlaoui H, Delbauve S, Passon N, 55. Ostrop J, Jozefowski K, Zimmermann S, Hofmann K, Strasser E,
 Twyffels L, Leo O, Flamand V: Neonatal follicular Th cell Lepenies B, Lang R: Contribution of MINCLE-SYK signaling to
responses are impaired and modulated by IL-4. J Immunol activation of primary human APCs by mycobacterial cord factor
2013, 191:1231-1239. and the novel adjuvant TDB. J Immunol 2015, 195:2417-2428.
See annotation to Ref. [35]. 56. Kamath AT, Mastelic B, Christensen D, Rochat AF, Agger EM,
38. Kool M, Fierens K, Lambrecht BN: Alum adjuvant: some of the Pinschewer DD, Andersen P, Lambert PH, Siegrist CA:
tricks of the oldest adjuvant. J Med Microbiol 2012, 61:927-934. Synchronization of dendritic cell activation and antigen
exposure is required for the induction of Th1/Th17 responses.
39. He Y, Zeng MY, Yang D, Motro B, Nunez G: NEK7 is an essential J Immunol 2012, 188:4828-4837.
mediator of NLRP3 activation downstream of potassium
efflux. Nature 2016, 530:354-357. 57. Kamath AT, Rochat AF, Christensen D, Agger EM, Andersen P,
Lambert PH, Siegrist CA: A liposome-based mycobacterial
40. Shi H, Wang Y, Li X, Zhan X, Tang M, Fina M, Su L, Pratt D, Bu CH, vaccine induces potent adult and neonatal multifunctional T
Hildebrand S et al.: NLRP3 activation and mitosis are mutually cells through the exquisite targeting of dendritic cells. PLoS
exclusive events coordinated by NEK7, a new inflammasome ONE 2009, 4:e5771.
component. Nat Immunol 2016, 17:250-258.
58. Agger EM, Rosenkrands I, Hansen J, Brahimi K, Vandahl BS,
41. Schallert N, Pihlgren M, Kovarik J, Roduit C, Tougne C, Bozzotti P, Aagaard C, Werninghaus K, Kirschning C, Lang R, Christensen D
Del Giudice G, Siegrist CA, Lambert PH: Generation of adult-like et al.: Cationic liposomes formulated with synthetic

www.sciencedirect.com Current Opinion in Immunology 2016, 41:1–8


8 Vaccines

mycobacterial cordfactor (CAF01): a versatile adjuvant for 64. Honda-Okubo Y, Ong CH, Petrovsky N: Advax delta inulin
vaccines with different immunological requirements. PLoS adjuvant overcomes immune immaturity in neonatal mice
ONE 2008, 3:e3116. thereby allowing single-dose influenza vaccine protection.
Vaccine 2015, 33:4892-4900.
59. van Dissel JT, Joosten SA, Hoff ST, Soonawala D, Prins C,
Hokey DA, O’Dee DM, Graves A, Thierry-Carstensen B, 65. Stassijns J, Bollaerts K, Baay M, Verstraeten T: A systematic
Andreasen LV et al.: A novel liposomal adjuvant system, CAF01,  review and meta-analysis on the safety of newly adjuvanted
promotes long-lived Mycobacterium tuberculosis-specific vaccines among children. Vaccine 2016, 34:714-722.
T-cell responses in human. Vaccine 2014, 32:7098-7107. This analysis is a state-of-the-art review of the latest developments and
trials for adjuvants in children.
60. Ota MO, Vekemans J, Schlegel-Haueter SE, Fielding K, Sanneh M,
Kidd M, Newport MJ, Aaby P, Whittle H, Lambert PH et al.: 66. Madhi SA, Cutland CL, Kuwanda L, Weinberg A, Hugo A, Jones S,
Influence of Mycobacterium bovis bacillus Calmette-Guerin  Adrian PV, van Niekerk N, Treurnicht F, Ortiz JR et al.: Influenza
on antibody and cytokine responses to human neonatal vaccination of pregnant women and protection of their infants.
vaccination. J Immunol 2002, 168:919-925. N Engl J Med 2014, 371:918-931.
These two studies show that maternal immunization may confer protec-
61. Vesikari T, Knuf M, Wutzler P, Karvonen A, Kieninger-Baum D,
tion through the effective transfer of protective maternal antibodies,
Schmitt HJ, Baehner F, Borkowski A, Tsai TF, Clemens R: Oil-in-
proving maternal immunization as currently the most effective strategy
water emulsion adjuvant with influenza vaccine in young
for neonatal protection.
children. N Engl J Med 2011, 365:1406-1416.
62. Nakaya HI, Clutterbuck E, Kazmin D, Wang L, Cortese M, 67. Amirthalingam G, Andrews N, Campbell H, Ribeiro S, Kara E,
Bosinger SE, Patel NB, Zak DE, Aderem A, Dong T et al.: Systems  Donegan K, Fry NK, Miller E, Ramsay M: Effectiveness of
biology of immunity to MF59-adjuvanted versus maternal pertussis vaccination in England: an observational
nonadjuvanted trivalent seasonal influenza vaccines in early study. Lancet 2014, 384:1521-1528.
childhood. Proc Natl Acad Sci U S A 2016. These two studies show that maternal immunization may confer protec-
tion through the effective transfer of protective maternal antibodies,
63. Vono M, Taccone M, Caccin P, Gallotta M, Donvito G, Falzoni S, proving maternal immunization as currently the most effective strategy
 Palmieri E, Pallaoro M, Rappuoli R, Di Virgilio F et al.: The adjuvant for neonatal protection.
MF59 induces ATP release from muscle that potentiates
response to vaccination. Proc Natl Acad Sci U S A 2013, 68. Dowling DJ, Tan Z, Prokopowicz ZM, Palmer CD, Matthews MA,
110:21095-21100. Dietsch GN, Hershberg RM, Levy O: The ultra-potent and
This study uncovers a novel mechanism relying on ATP release at the site selective TLR8 agonist VTX-294 activates human newborn and
of injection whereby MF59 adjuvantation promotes the recruitment of adult leukocytes. PLoS ONE 2013, 8:e58164.
innate cells.

Current Opinion in Immunology 2016, 41:1–8 www.sciencedirect.com

You might also like