Skip to main content
Top
Published in: Seminars in Immunopathology 6/2017

Open Access 01-11-2017 | Review

Vaccine responses in newborns

Authors: Anja Saso, Beate Kampmann

Published in: Seminars in Immunopathology | Issue 6/2017

Login to get access

Abstract

Immunisation of the newborn represents a key global strategy in overcoming morbidity and mortality due to infection in early life. Potential limitations, however, include poor immunogenicity, safety concerns and the development of tolerogenicity or hypo-responsiveness to either the same antigen and/or concomitant antigens administered at birth or in the subsequent months. Furthermore, the neonatal immunological milieu is polarised towards Th2-type immunity with dampening of Th1-type responses and impaired humoral immunity, resulting in qualitatively and quantitatively poorer antibody responses compared to older infants. Innate immunity also shows functional deficiency in antigen-presenting cells: the expression and signalling of Toll-like receptors undergo maturational changes associated with distinct functional responses. Nevertheless, the effectiveness of BCG, hepatitis B and oral polio vaccines, the only immunisations currently in use in the neonatal period, is proof of concept that vaccines can be successfully administered to the newborn via different routes of delivery to induce a range of protective mechanisms for three different diseases. In this review paper, we discuss the rationale for and challenges to neonatal immunisation, summarising progress made in the field, including lessons learnt from newborn vaccines in the pipeline. Furthermore, we explore important maternal, infant and environmental co-factors that may impede the success of current and future neonatal immunisation strategies. A variety of approaches have been proposed to overcome the inherent regulatory constraints of the newborn innate and adaptive immune system, including alternative routes of delivery, novel vaccine configurations, improved innate receptor agonists and optimised antigen-adjuvant combinations. Crucially, a dual strategy may be employed whereby immunisation at birth is used to prime the immune system in order to improve immunogenicity to subsequent homologous or heterologous boosters in later infancy. Similarly, potent non-specific immunomodulatory effects may be elicited when challenged with unrelated antigens, with the potential to reduce the overall risk of infection and allergic disease in early life.
Literature
2.
go back to reference Kollmann TR, Kampmann B, Mazmanian SK, Marchant A, Levy O (2017) Protecting the newborn and young infant from infectious diseases: lessons from immune ontogeny. Immunity 46:350–363PubMedCrossRef Kollmann TR, Kampmann B, Mazmanian SK, Marchant A, Levy O (2017) Protecting the newborn and young infant from infectious diseases: lessons from immune ontogeny. Immunity 46:350–363PubMedCrossRef
3.
go back to reference Wood N, Siegrist CA (2011) Neonatal immunization: where do we stand? Curr Opin Infect Dis 24:190–195PubMedCrossRef Wood N, Siegrist CA (2011) Neonatal immunization: where do we stand? Curr Opin Infect Dis 24:190–195PubMedCrossRef
8.
go back to reference Goriely S, Goldman M (2007) From tolerance to autoimmunity: is there a risk in early life vaccination? J Comp Pathol 137(Suppl 1):S57–S61PubMedCrossRef Goriely S, Goldman M (2007) From tolerance to autoimmunity: is there a risk in early life vaccination? J Comp Pathol 137(Suppl 1):S57–S61PubMedCrossRef
11.
12.
go back to reference Garcia AM, Fadel SA, Cao S, Sarzotti M (2000) T cell immunity in neonates. Immunol Res 22:177–190PubMedCrossRef Garcia AM, Fadel SA, Cao S, Sarzotti M (2000) T cell immunity in neonates. Immunol Res 22:177–190PubMedCrossRef
15.
go back to reference PrabhuDas M, Adkins B, Gans H, King C, Levy O, Ramilo O, Siegrist CA (2011) Challenges in infant immunity: implications for responses to infection and vaccines. Nat Immunol 12:189–194PubMedCrossRef PrabhuDas M, Adkins B, Gans H, King C, Levy O, Ramilo O, Siegrist CA (2011) Challenges in infant immunity: implications for responses to infection and vaccines. Nat Immunol 12:189–194PubMedCrossRef
16.
go back to reference Siegrist CA (2007) The challenges of vaccine responses in early life: selected examples. J Comp Pathol 137(Suppl 1):S4–S9PubMedCrossRef Siegrist CA (2007) The challenges of vaccine responses in early life: selected examples. J Comp Pathol 137(Suppl 1):S4–S9PubMedCrossRef
18.
go back to reference Haines CJ, Giffon TD, Lu LS, Lu X, Tessier-Lavigne M, Ross DT, Lewis DB (2009) Human CD4+ T cell recent thymic emigrants are identified by protein tyrosine kinase 7 and have reduced immune function. J Exp Med 206:275–285PubMedPubMedCentralCrossRef Haines CJ, Giffon TD, Lu LS, Lu X, Tessier-Lavigne M, Ross DT, Lewis DB (2009) Human CD4+ T cell recent thymic emigrants are identified by protein tyrosine kinase 7 and have reduced immune function. J Exp Med 206:275–285PubMedPubMedCentralCrossRef
19.
go back to reference White GP, Watt PM, Holt BJ, Holt PG (2002) Differential patterns of methylation of the IFN-gamma promoter at CpG and non-CpG sites underlie differences in IFN-gamma gene expression between human neonatal and adult CD45RO- T cells. J Immunol 168:2820–2827PubMedCrossRef White GP, Watt PM, Holt BJ, Holt PG (2002) Differential patterns of methylation of the IFN-gamma promoter at CpG and non-CpG sites underlie differences in IFN-gamma gene expression between human neonatal and adult CD45RO- T cells. J Immunol 168:2820–2827PubMedCrossRef
20.
go back to reference Ma CS, Suryani S, Avery DT, Chan A, Nanan R, Santner-Nanan B, Deenick EK, Tangye SG (2009) Early commitment of naive human CD4(+) T cells to the T follicular helper (T(FH)) cell lineage is induced by IL-12. Immunol Cell Biol 87:590–600PubMedCrossRef Ma CS, Suryani S, Avery DT, Chan A, Nanan R, Santner-Nanan B, Deenick EK, Tangye SG (2009) Early commitment of naive human CD4(+) T cells to the T follicular helper (T(FH)) cell lineage is induced by IL-12. Immunol Cell Biol 87:590–600PubMedCrossRef
21.
22.
go back to reference Boer MC, Joosten SA, Ottenhoff TH (2015) Regulatory T-cells at the interface between human host and pathogens in infectious diseases and vaccination. Front Immunol 6:217PubMedPubMedCentralCrossRef Boer MC, Joosten SA, Ottenhoff TH (2015) Regulatory T-cells at the interface between human host and pathogens in infectious diseases and vaccination. Front Immunol 6:217PubMedPubMedCentralCrossRef
23.
go back to reference Burt TD (2013) Fetal regulatory T cells and peripheral immune tolerance in utero: implications for development and disease. Am J Reprod Immunol 69:346–358PubMedPubMedCentralCrossRef Burt TD (2013) Fetal regulatory T cells and peripheral immune tolerance in utero: implications for development and disease. Am J Reprod Immunol 69:346–358PubMedPubMedCentralCrossRef
24.
go back to reference Kollmann TR, Levy O, Montgomery RR, Goriely S (2012) Innate immune function by Toll-like receptors: distinct responses in newborns and the elderly. Immunity 37:771–783PubMedPubMedCentralCrossRef Kollmann TR, Levy O, Montgomery RR, Goriely S (2012) Innate immune function by Toll-like receptors: distinct responses in newborns and the elderly. Immunity 37:771–783PubMedPubMedCentralCrossRef
25.
go back to reference Siegrist CA, Aspinall R (2009) B-cell responses to vaccination at the extremes of age. Nat Rev Immunol 9:185–194PubMedCrossRef Siegrist CA, Aspinall R (2009) B-cell responses to vaccination at the extremes of age. Nat Rev Immunol 9:185–194PubMedCrossRef
26.
go back to reference Debock I, Jaworski K, Chadlaoui H, Delbauve S, Passon N, Twyffels L, Leo O, Flamand V (2013) Neonatal follicular Th cell responses are impaired and modulated by IL-4. J Immunol 191:1231–1239PubMedCrossRef Debock I, Jaworski K, Chadlaoui H, Delbauve S, Passon N, Twyffels L, Leo O, Flamand V (2013) Neonatal follicular Th cell responses are impaired and modulated by IL-4. J Immunol 191:1231–1239PubMedCrossRef
27.
go back to reference Mastelic B, Kamath AT, Fontannaz P, Tougne C, Rochat AF, Belnoue E, Combescure C, Auderset F, Lambert PH, Tacchini-Cottier F, Siegrist CA (2012) Environmental and T cell-intrinsic factors limit the expansion of neonatal follicular T helper cells but may be circumvented by specific adjuvants. J Immunol 189:5764–5772PubMedCrossRef Mastelic B, Kamath AT, Fontannaz P, Tougne C, Rochat AF, Belnoue E, Combescure C, Auderset F, Lambert PH, Tacchini-Cottier F, Siegrist CA (2012) Environmental and T cell-intrinsic factors limit the expansion of neonatal follicular T helper cells but may be circumvented by specific adjuvants. J Immunol 189:5764–5772PubMedCrossRef
28.
go back to reference Pihlgren M, Tougne C, Bozzotti P, Fulurija A, Duchosal MA, Lambert PH, Siegrist CA (2003) Unresponsiveness to lymphoid-mediated signals at the neonatal follicular dendritic cell precursor level contributes to delayed germinal center induction and limitations of neonatal antibody responses to T-dependent antigens. J Immunol 170:2824–2832PubMedCrossRef Pihlgren M, Tougne C, Bozzotti P, Fulurija A, Duchosal MA, Lambert PH, Siegrist CA (2003) Unresponsiveness to lymphoid-mediated signals at the neonatal follicular dendritic cell precursor level contributes to delayed germinal center induction and limitations of neonatal antibody responses to T-dependent antigens. J Immunol 170:2824–2832PubMedCrossRef
29.
go back to reference Mastelic Gavillet B, Eberhardt CS, Auderset F, Castellino F, Seubert A, Tregoning JS, Lambert PH, de Gregorio E, Del Giudice G, Siegrist CA (2015) MF59 mediates its B cell adjuvanticity by promoting T follicular helper cells and thus germinal center responses in adult and early life. J Immunol 194:4836–4845PubMedCrossRef Mastelic Gavillet B, Eberhardt CS, Auderset F, Castellino F, Seubert A, Tregoning JS, Lambert PH, de Gregorio E, Del Giudice G, Siegrist CA (2015) MF59 mediates its B cell adjuvanticity by promoting T follicular helper cells and thus germinal center responses in adult and early life. J Immunol 194:4836–4845PubMedCrossRef
30.
go back to reference Levy O, Coughlin M, Cronstein BN, Roy RM, Desai A, Wessels MR (2006) The adenosine system selectively inhibits TLR-mediated TNF-alpha production in the human newborn. J Immunol 177:1956–1966PubMedPubMedCentralCrossRef Levy O, Coughlin M, Cronstein BN, Roy RM, Desai A, Wessels MR (2006) The adenosine system selectively inhibits TLR-mediated TNF-alpha production in the human newborn. J Immunol 177:1956–1966PubMedPubMedCentralCrossRef
31.
go back to reference Levy O (2007) Innate immunity of the newborn: basic mechanisms and clinical correlates. Nat Rev Immunol 7:379–390PubMedCrossRef Levy O (2007) Innate immunity of the newborn: basic mechanisms and clinical correlates. Nat Rev Immunol 7:379–390PubMedCrossRef
33.
go back to reference Kampmann B, Jones CE (2015) Factors influencing innate immunity and vaccine responses in infancy. Philos Trans R Soc Lond Ser B Biol Sci 19;370(1671) Kampmann B, Jones CE (2015) Factors influencing innate immunity and vaccine responses in infancy. Philos Trans R Soc Lond Ser B Biol Sci 19;370(1671)
34.
go back to reference Corbett NP, Blimkie D, Ho KC, Cai B, Sutherland DP, Kallos A, Crabtree J, Rein-Weston A, Lavoie PM, Turvey SE, Hawkins NR, Self SG, Wilson CB, Hajjar AM, Fortuno ES 3rd, Kollmann TR (2010) Ontogeny of Toll-like receptor mediated cytokine responses of human blood mononuclear cells. PLoS One 5:e15041PubMedPubMedCentralCrossRef Corbett NP, Blimkie D, Ho KC, Cai B, Sutherland DP, Kallos A, Crabtree J, Rein-Weston A, Lavoie PM, Turvey SE, Hawkins NR, Self SG, Wilson CB, Hajjar AM, Fortuno ES 3rd, Kollmann TR (2010) Ontogeny of Toll-like receptor mediated cytokine responses of human blood mononuclear cells. PLoS One 5:e15041PubMedPubMedCentralCrossRef
35.
go back to reference Levy O (2005) Innate immunity of the human newborn: distinct cytokine responses to LPS and other Toll-like receptor agonists. J Endotoxin Res 11:113–116PubMedCrossRef Levy O (2005) Innate immunity of the human newborn: distinct cytokine responses to LPS and other Toll-like receptor agonists. J Endotoxin Res 11:113–116PubMedCrossRef
36.
go back to reference Ota MO, Vekemans J, Schlegel-Haueter SE, Fielding K, Whittle H, Lambert PH, McAdam KP, Siegrist CA, Marchant A (2004) Hepatitis B immunisation induces higher antibody and memory Th2 responses in new-borns than in adults. Vaccine 22:511–519PubMedCrossRef Ota MO, Vekemans J, Schlegel-Haueter SE, Fielding K, Whittle H, Lambert PH, McAdam KP, Siegrist CA, Marchant A (2004) Hepatitis B immunisation induces higher antibody and memory Th2 responses in new-borns than in adults. Vaccine 22:511–519PubMedCrossRef
37.
go back to reference Vekemans J, Ota MO, Wang EC, Kidd M, Borysiewicz LK, Whittle H, McAdam KP, Morgan G, Marchant A (2002) T cell responses to vaccines in infants: defective IFNgamma production after oral polio vaccination. Clin Exp Immunol 127:495–498PubMedPubMedCentralCrossRef Vekemans J, Ota MO, Wang EC, Kidd M, Borysiewicz LK, Whittle H, McAdam KP, Morgan G, Marchant A (2002) T cell responses to vaccines in infants: defective IFNgamma production after oral polio vaccination. Clin Exp Immunol 127:495–498PubMedPubMedCentralCrossRef
38.
go back to reference Ota MO, Vekemans J, Schlegel-Haueter SE, Fielding K, Sanneh M, Kidd M, Newport MJ, Aaby P, Whittle H, Lambert PH, McAdam KP, Siegrist CA, Marchant A (2002) Influence of Mycobacterium bovis bacillus Calmette-Guerin on antibody and cytokine responses to human neonatal vaccination. J Immunol 168:919–925PubMedCrossRef Ota MO, Vekemans J, Schlegel-Haueter SE, Fielding K, Sanneh M, Kidd M, Newport MJ, Aaby P, Whittle H, Lambert PH, McAdam KP, Siegrist CA, Marchant A (2002) Influence of Mycobacterium bovis bacillus Calmette-Guerin on antibody and cytokine responses to human neonatal vaccination. J Immunol 168:919–925PubMedCrossRef
39.
go back to reference Siegrist CA (2008) Blame vaccine interference, not neonatal immunization, for suboptimal responses after neonatal diphtheria, tetanus, and acellular pertussis immunization. J Pediatr 153:305–307PubMedCrossRef Siegrist CA (2008) Blame vaccine interference, not neonatal immunization, for suboptimal responses after neonatal diphtheria, tetanus, and acellular pertussis immunization. J Pediatr 153:305–307PubMedCrossRef
40.
go back to reference Halasa NB, O'Shea A, Shi JR, LaFleur BJ, Edwards KM (2008) Poor immune responses to a birth dose of diphtheria, tetanus, and acellular pertussis vaccine. J Pediatr 153:327–332PubMedPubMedCentralCrossRef Halasa NB, O'Shea A, Shi JR, LaFleur BJ, Edwards KM (2008) Poor immune responses to a birth dose of diphtheria, tetanus, and acellular pertussis vaccine. J Pediatr 153:327–332PubMedPubMedCentralCrossRef
41.
go back to reference Knuf M, Schmitt HJ, Wolter J, Schuerman L, Jacquet JM, Kieninger D, Siegrist CA, Zepp F (2008) Neonatal vaccination with an acellular pertussis vaccine accelerates the acquisition of pertussis antibodies in infants. J Pediatr 152:655–660 60.e1PubMedCrossRef Knuf M, Schmitt HJ, Wolter J, Schuerman L, Jacquet JM, Kieninger D, Siegrist CA, Zepp F (2008) Neonatal vaccination with an acellular pertussis vaccine accelerates the acquisition of pertussis antibodies in infants. J Pediatr 152:655–660 60.e1PubMedCrossRef
42.
go back to reference Knuf M, Schmitt HJ, Jacquet JM, Collard A, Kieninger D, Meyer CU, Siegrist CA, Zepp F (2010) Booster vaccination after neonatal priming with acellular pertussis vaccine. J Pediatr 156:675–678PubMedCrossRef Knuf M, Schmitt HJ, Jacquet JM, Collard A, Kieninger D, Meyer CU, Siegrist CA, Zepp F (2010) Booster vaccination after neonatal priming with acellular pertussis vaccine. J Pediatr 156:675–678PubMedCrossRef
43.
go back to reference Nissen TN, Birk NM, Smits G, Jeppesen DL, Stensballe LG, Netea MG, van der Klis F, Benn CS, Pryds O (2017) Bacille Calmette-Guerin (BCG) vaccination at birth and antibody responses to childhood vaccines. A randomised clinical trial. Vaccine 35:2084–2091PubMedCrossRef Nissen TN, Birk NM, Smits G, Jeppesen DL, Stensballe LG, Netea MG, van der Klis F, Benn CS, Pryds O (2017) Bacille Calmette-Guerin (BCG) vaccination at birth and antibody responses to childhood vaccines. A randomised clinical trial. Vaccine 35:2084–2091PubMedCrossRef
44.
go back to reference Halsey N, Galazka A (1985) The efficacy of DPT and oral poliomyelitis immunization schedules initiated from birth to 12 weeks of age. Bull World Health Organ 63:1151–1169PubMedPubMedCentral Halsey N, Galazka A (1985) The efficacy of DPT and oral poliomyelitis immunization schedules initiated from birth to 12 weeks of age. Bull World Health Organ 63:1151–1169PubMedPubMedCentral
45.
go back to reference Trunz BB, Fine P, Dye C (2006) Effect of BCG vaccination on childhood tuberculous meningitis and miliary tuberculosis worldwide: a meta-analysis and assessment of cost-effectiveness. Lancet 367:1173–1180PubMedCrossRef Trunz BB, Fine P, Dye C (2006) Effect of BCG vaccination on childhood tuberculous meningitis and miliary tuberculosis worldwide: a meta-analysis and assessment of cost-effectiveness. Lancet 367:1173–1180PubMedCrossRef
46.
go back to reference Roy A, Eisenhut M, Harris RJ, Rodrigues LC, Sridhar S, Habermann S, Snell L, Mangtani P, Adetifa I, Lalvani A, Abubakar I (2014) Effect of BCG vaccination against Mycobacterium tuberculosis infection in children: systematic review and meta-analysis. BMJ 349:g4643PubMedPubMedCentralCrossRef Roy A, Eisenhut M, Harris RJ, Rodrigues LC, Sridhar S, Habermann S, Snell L, Mangtani P, Adetifa I, Lalvani A, Abubakar I (2014) Effect of BCG vaccination against Mycobacterium tuberculosis infection in children: systematic review and meta-analysis. BMJ 349:g4643PubMedPubMedCentralCrossRef
47.
48.
go back to reference Clemens JD, Chuong JJ, Feinstein AR (1983) The BCG controversy. A methodological and statistical reappraisal. JAMA 249:2362–2369PubMedCrossRef Clemens JD, Chuong JJ, Feinstein AR (1983) The BCG controversy. A methodological and statistical reappraisal. JAMA 249:2362–2369PubMedCrossRef
49.
go back to reference Colditz GA, Berkey CS, Mosteller F, Brewer TF, Wilson ME, Burdick E, Fineberg HV (1995) The efficacy of bacillus Calmette-Guerin vaccination of newborns and infants in the prevention of tuberculosis: meta-analyses of the published literature. Pediatrics 96:29–35PubMed Colditz GA, Berkey CS, Mosteller F, Brewer TF, Wilson ME, Burdick E, Fineberg HV (1995) The efficacy of bacillus Calmette-Guerin vaccination of newborns and infants in the prevention of tuberculosis: meta-analyses of the published literature. Pediatrics 96:29–35PubMed
50.
go back to reference Remus N, Reichenbach J, Picard C, Rietschel C, Wood P, Lammas D, Kumararatne DS, Casanova JL (2001) Impaired interferon gamma-mediated immunity and susceptibility to mycobacterial infection in childhood. Pediatr Res 50:8–13PubMedCrossRef Remus N, Reichenbach J, Picard C, Rietschel C, Wood P, Lammas D, Kumararatne DS, Casanova JL (2001) Impaired interferon gamma-mediated immunity and susceptibility to mycobacterial infection in childhood. Pediatr Res 50:8–13PubMedCrossRef
51.
go back to reference Fine PE (1995) Variation in protection by BCG: implications of and for heterologous immunity. Lancet 346:1339–1345PubMedCrossRef Fine PE (1995) Variation in protection by BCG: implications of and for heterologous immunity. Lancet 346:1339–1345PubMedCrossRef
52.
go back to reference Heldwein KA, Liang MD, Andresen TK, Thomas KE, Marty AM, Cuesta N, Vogel SN, Fenton MJ (2003) TLR2 and TLR4 serve distinct roles in the host immune response against Mycobacterium bovis BCG. J Leukoc Biol 74:277–286PubMedCrossRef Heldwein KA, Liang MD, Andresen TK, Thomas KE, Marty AM, Cuesta N, Vogel SN, Fenton MJ (2003) TLR2 and TLR4 serve distinct roles in the host immune response against Mycobacterium bovis BCG. J Leukoc Biol 74:277–286PubMedCrossRef
53.
go back to reference Mangtani P, Abubakar I, Ariti C, Beynon R, Pimpin L, Fine PE, Rodrigues LC, Smith PG, Lipman M, Whiting PF, Sterne JA (2014) Protection by BCG vaccine against tuberculosis: a systematic review of randomized controlled trials. Clin Infect Dis 58:470–480PubMedCrossRef Mangtani P, Abubakar I, Ariti C, Beynon R, Pimpin L, Fine PE, Rodrigues LC, Smith PG, Lipman M, Whiting PF, Sterne JA (2014) Protection by BCG vaccine against tuberculosis: a systematic review of randomized controlled trials. Clin Infect Dis 58:470–480PubMedCrossRef
54.
go back to reference Marchant A, Goetghebuer T, Ota MO, Wolfe I, Ceesay SJ, De Groote D, Corrah T, Bennett S, Wheeler J, Huygen K, Aaby P, McAdam KP, Newport MJ (1999) Newborns develop a Th1-type immune response to Mycobacterium bovis bacillus Calmette-Guerin vaccination. J Immunol 163:2249–2255PubMed Marchant A, Goetghebuer T, Ota MO, Wolfe I, Ceesay SJ, De Groote D, Corrah T, Bennett S, Wheeler J, Huygen K, Aaby P, McAdam KP, Newport MJ (1999) Newborns develop a Th1-type immune response to Mycobacterium bovis bacillus Calmette-Guerin vaccination. J Immunol 163:2249–2255PubMed
55.
go back to reference Soares AP, Kwong Chung CK, Choice T, Hughes EJ, Jacobs G, van Rensburg EJ, Khomba G, de Kock M, Lerumo L, Makhethe L, Maneli MH, Pienaar B, Smit E, Tena-Coki NG, van Wyk L, Boom WH, Kaplan G, Scriba TJ, Hanekom WA (2013) Longitudinal changes in CD4(+) T-cell memory responses induced by BCG vaccination of newborns. J Infect Dis 207:1084–1094PubMedPubMedCentralCrossRef Soares AP, Kwong Chung CK, Choice T, Hughes EJ, Jacobs G, van Rensburg EJ, Khomba G, de Kock M, Lerumo L, Makhethe L, Maneli MH, Pienaar B, Smit E, Tena-Coki NG, van Wyk L, Boom WH, Kaplan G, Scriba TJ, Hanekom WA (2013) Longitudinal changes in CD4(+) T-cell memory responses induced by BCG vaccination of newborns. J Infect Dis 207:1084–1094PubMedPubMedCentralCrossRef
56.
go back to reference Kagina BM, Abel B, Scriba TJ, Hughes EJ, Keyser A, Soares A, Gamieldien H, Sidibana M, Hatherill M, Gelderbloem S, Mahomed H, Hawkridge A, Hussey G, Kaplan G, Hanekom WA (2010) Specific T cell frequency and cytokine expression profile do not correlate with protection against tuberculosis after bacillus Calmette-Guerin vaccination of newborns. Am J Respir Crit Care Med 182:1073–1079PubMedPubMedCentralCrossRef Kagina BM, Abel B, Scriba TJ, Hughes EJ, Keyser A, Soares A, Gamieldien H, Sidibana M, Hatherill M, Gelderbloem S, Mahomed H, Hawkridge A, Hussey G, Kaplan G, Hanekom WA (2010) Specific T cell frequency and cytokine expression profile do not correlate with protection against tuberculosis after bacillus Calmette-Guerin vaccination of newborns. Am J Respir Crit Care Med 182:1073–1079PubMedPubMedCentralCrossRef
57.
go back to reference Aaby P, Benn CS (2011) Non-specific and sex-differential effects of routine vaccines: what evidence is needed to take these effects into consideration in low-income countries? Hum Vaccin 7:120–124PubMedCrossRef Aaby P, Benn CS (2011) Non-specific and sex-differential effects of routine vaccines: what evidence is needed to take these effects into consideration in low-income countries? Hum Vaccin 7:120–124PubMedCrossRef
58.
go back to reference Cavallo GP, Elia M, Giordano D, Baldi C, Cammarota R (2002) Decrease of specific and total IgE levels in allergic patients after BCG vaccination: preliminary report. Arch Otolaryngol Head Neck Surg 128:1058–1060PubMedCrossRef Cavallo GP, Elia M, Giordano D, Baldi C, Cammarota R (2002) Decrease of specific and total IgE levels in allergic patients after BCG vaccination: preliminary report. Arch Otolaryngol Head Neck Surg 128:1058–1060PubMedCrossRef
59.
go back to reference de Castro MJ, Pardo-Seco J, Martinon-Torres F (2015) Nonspecific (heterologous) protection of neonatal BCG vaccination against hospitalization due to respiratory infection and sepsis. Clin Infect Dis 60:1611–1619PubMedCrossRef de Castro MJ, Pardo-Seco J, Martinon-Torres F (2015) Nonspecific (heterologous) protection of neonatal BCG vaccination against hospitalization due to respiratory infection and sepsis. Clin Infect Dis 60:1611–1619PubMedCrossRef
60.
go back to reference Freyne B, Marchant A, Curtis N (2015) BCG-associated heterologous immunity, a historical perspective: intervention studies in animal models of infectious diseases. Trans R Soc Trop Med Hyg 109:52–61PubMedCrossRef Freyne B, Marchant A, Curtis N (2015) BCG-associated heterologous immunity, a historical perspective: intervention studies in animal models of infectious diseases. Trans R Soc Trop Med Hyg 109:52–61PubMedCrossRef
61.
go back to reference Garly ML, Martins CL, Bale C, Balde MA, Hedegaard KL, Gustafson P, Lisse IM, Whittle HC, Aaby P (2003) BCG scar and positive tuberculin reaction associated with reduced child mortality in West Africa. A non-specific beneficial effect of BCG? Vaccine 21:2782–2790PubMedCrossRef Garly ML, Martins CL, Bale C, Balde MA, Hedegaard KL, Gustafson P, Lisse IM, Whittle HC, Aaby P (2003) BCG scar and positive tuberculin reaction associated with reduced child mortality in West Africa. A non-specific beneficial effect of BCG? Vaccine 21:2782–2790PubMedCrossRef
62.
go back to reference Goodridge HS, Ahmed SS, Curtis N, Kollmann TR, Levy O, Netea MG, Pollard AJ, van Crevel R, Wilson CB (2016) Harnessing the beneficial heterologous effects of vaccination. Nat Rev Immunol 16:392–400PubMedPubMedCentralCrossRef Goodridge HS, Ahmed SS, Curtis N, Kollmann TR, Levy O, Netea MG, Pollard AJ, van Crevel R, Wilson CB (2016) Harnessing the beneficial heterologous effects of vaccination. Nat Rev Immunol 16:392–400PubMedPubMedCentralCrossRef
63.
go back to reference Fine PE, Smith PG, Evans SJ (2012) Non-specific effects of BCG? J Infect Dis 205:515 author reply 7-8 PubMedCrossRef Fine PE, Smith PG, Evans SJ (2012) Non-specific effects of BCG? J Infect Dis 205:515 author reply 7-8 PubMedCrossRef
64.
go back to reference Stensballe LG, Sorup S, Aaby P, Benn CS, Greisen G, Jeppesen DL, Birk NM, Kjaergaard J, Nissen TN, Pihl GT, Thostesen LM, Kofoed PE, Pryds O, Ravn H (2017) BCG vaccination at birth and early childhood hospitalisation: a randomised clinical multicentre trial. Arch Dis Child 102:224–231PubMedCrossRef Stensballe LG, Sorup S, Aaby P, Benn CS, Greisen G, Jeppesen DL, Birk NM, Kjaergaard J, Nissen TN, Pihl GT, Thostesen LM, Kofoed PE, Pryds O, Ravn H (2017) BCG vaccination at birth and early childhood hospitalisation: a randomised clinical multicentre trial. Arch Dis Child 102:224–231PubMedCrossRef
65.
go back to reference Kandasamy R, Voysey M, McQuaid F, de Nie K, Ryan R, Orr O, Uhlig U, Sande C, O'Connor D, Pollard AJ (2016) Non-specific immunological effects of selected routine childhood immunisations: systematic review. BMJ 355:i5225PubMedPubMedCentralCrossRef Kandasamy R, Voysey M, McQuaid F, de Nie K, Ryan R, Orr O, Uhlig U, Sande C, O'Connor D, Pollard AJ (2016) Non-specific immunological effects of selected routine childhood immunisations: systematic review. BMJ 355:i5225PubMedPubMedCentralCrossRef
66.
go back to reference Pollard AJ, Finn A, Curtis N (2017) Non-specific effects of vaccines: plausible and potentially important, but implications uncertain. Arch Dis Child Pollard AJ, Finn A, Curtis N (2017) Non-specific effects of vaccines: plausible and potentially important, but implications uncertain. Arch Dis Child
67.
go back to reference Ritz N, Mui M, Balloch A, Curtis N (2013) Non-specific effect of Bacille Calmette-Guerin vaccine on the immune response to routine immunisations. Vaccine 31:3098–3103PubMedCrossRef Ritz N, Mui M, Balloch A, Curtis N (2013) Non-specific effect of Bacille Calmette-Guerin vaccine on the immune response to routine immunisations. Vaccine 31:3098–3103PubMedCrossRef
68.
go back to reference Ifrim DC, Quintin J, Joosten LA, Jacobs C, Jansen T, Jacobs L, Gow NA, Williams DL, van der Meer JW, Netea MG (2014) Trained immunity or tolerance: opposing functional programs induced in human monocytes after engagement of various pattern recognition receptors. Clin Vaccine Immunol 21:534–545PubMedPubMedCentralCrossRef Ifrim DC, Quintin J, Joosten LA, Jacobs C, Jansen T, Jacobs L, Gow NA, Williams DL, van der Meer JW, Netea MG (2014) Trained immunity or tolerance: opposing functional programs induced in human monocytes after engagement of various pattern recognition receptors. Clin Vaccine Immunol 21:534–545PubMedPubMedCentralCrossRef
70.
go back to reference Levy O, Netea MG (2014) Innate immune memory: implications for development of pediatric immunomodulatory agents and adjuvanted vaccines. Pediatr Res 75:184–188PubMedCrossRef Levy O, Netea MG (2014) Innate immune memory: implications for development of pediatric immunomodulatory agents and adjuvanted vaccines. Pediatr Res 75:184–188PubMedCrossRef
71.
go back to reference Kleinnijenhuis J, Quintin J, Preijers F, Joosten LA, Ifrim DC, Saeed S, Jacobs C, van Loenhout J, de Jong D, Stunnenberg HG, Xavier RJ, van der Meer JW, van Crevel R, Netea MG (2012) Bacille Calmette-Guerin induces NOD2-dependent nonspecific protection from reinfection via epigenetic reprogramming of monocytes. Proc Natl Acad Sci U S A 109:17537–17542PubMedPubMedCentralCrossRef Kleinnijenhuis J, Quintin J, Preijers F, Joosten LA, Ifrim DC, Saeed S, Jacobs C, van Loenhout J, de Jong D, Stunnenberg HG, Xavier RJ, van der Meer JW, van Crevel R, Netea MG (2012) Bacille Calmette-Guerin induces NOD2-dependent nonspecific protection from reinfection via epigenetic reprogramming of monocytes. Proc Natl Acad Sci U S A 109:17537–17542PubMedPubMedCentralCrossRef
72.
go back to reference Kleinnijenhuis J, Quintin J, Preijers F, Joosten LA, Jacobs C, Xavier RJ, van der Meer JW, van Crevel R, Netea MG (2014) BCG-induced trained immunity in NK cells: role for non-specific protection to infection. Clin Immunol 155:213–219PubMedPubMedCentralCrossRef Kleinnijenhuis J, Quintin J, Preijers F, Joosten LA, Jacobs C, Xavier RJ, van der Meer JW, van Crevel R, Netea MG (2014) BCG-induced trained immunity in NK cells: role for non-specific protection to infection. Clin Immunol 155:213–219PubMedPubMedCentralCrossRef
73.
go back to reference Djuardi Y, Sartono E, Wibowo H, Supali T, Yazdanbakhsh M (2010) A longitudinal study of BCG vaccination in early childhood: the development of innate and adaptive immune responses. PLoS One 5:e14066PubMedPubMedCentralCrossRef Djuardi Y, Sartono E, Wibowo H, Supali T, Yazdanbakhsh M (2010) A longitudinal study of BCG vaccination in early childhood: the development of innate and adaptive immune responses. PLoS One 5:e14066PubMedPubMedCentralCrossRef
74.
go back to reference Welsh RM, Selin LK (2002) No one is naive: the significance of heterologous T-cell immunity. Nat Rev Immunol 2:417–426PubMedCrossRef Welsh RM, Selin LK (2002) No one is naive: the significance of heterologous T-cell immunity. Nat Rev Immunol 2:417–426PubMedCrossRef
75.
go back to reference Gerlich WH (2015) Prophylactic vaccination against hepatitis B: achievements, challenges and perspectives. Med Microbiol Immunol 204:39–55PubMedCrossRef Gerlich WH (2015) Prophylactic vaccination against hepatitis B: achievements, challenges and perspectives. Med Microbiol Immunol 204:39–55PubMedCrossRef
76.
go back to reference Wong VC, Ip HM, Reesink HW, Lelie PN, Reerink-Brongers EE, Yeung CY, Ma HK (1984) Prevention of the HBsAg carrier state in newborn infants of mothers who are chronic carriers of HBsAg and HBeAg by administration of hepatitis-B vaccine and hepatitis-B immunoglobulin. Double-blind randomised placebo-controlled study. Lancet 1:921–926PubMedCrossRef Wong VC, Ip HM, Reesink HW, Lelie PN, Reerink-Brongers EE, Yeung CY, Ma HK (1984) Prevention of the HBsAg carrier state in newborn infants of mothers who are chronic carriers of HBsAg and HBeAg by administration of hepatitis-B vaccine and hepatitis-B immunoglobulin. Double-blind randomised placebo-controlled study. Lancet 1:921–926PubMedCrossRef
77.
go back to reference Xu ZY, Liu CB, Francis DP, Purcell RH, Gun ZL, Duan SC, Chen RJ, Margolis HS, Huang CH, Maynard JE (1985) Prevention of perinatal acquisition of hepatitis B virus carriage using vaccine: preliminary report of a randomized, double-blind placebo-controlled and comparative trial. Pediatrics 76:713–718PubMed Xu ZY, Liu CB, Francis DP, Purcell RH, Gun ZL, Duan SC, Chen RJ, Margolis HS, Huang CH, Maynard JE (1985) Prevention of perinatal acquisition of hepatitis B virus carriage using vaccine: preliminary report of a randomized, double-blind placebo-controlled and comparative trial. Pediatrics 76:713–718PubMed
79.
go back to reference Philbin VJ, Levy O (2007) Immunostimulatory activity of Toll-like receptor 8 agonists towards human leucocytes: basic mechanisms and translational opportunities. Biochem Soc Trans 35:1485–1491PubMedCrossRef Philbin VJ, Levy O (2007) Immunostimulatory activity of Toll-like receptor 8 agonists towards human leucocytes: basic mechanisms and translational opportunities. Biochem Soc Trans 35:1485–1491PubMedCrossRef
80.
go back to reference el-Sayed N, el-Gamal Y, Abbassy AA, Seoud I, Salama M, Kandeel A, Hossny E, Shawky A, Hussein HA, Pallansch MA, van der Avoort HG, Burton AH, Sreevatsava M, Malankar P, Wahdan MH, Sutter RW (2008) Monovalent type 1 oral poliovirus vaccine in newborns. N Engl J Med 359:1655–1665PubMedCrossRef el-Sayed N, el-Gamal Y, Abbassy AA, Seoud I, Salama M, Kandeel A, Hossny E, Shawky A, Hussein HA, Pallansch MA, van der Avoort HG, Burton AH, Sreevatsava M, Malankar P, Wahdan MH, Sutter RW (2008) Monovalent type 1 oral poliovirus vaccine in newborns. N Engl J Med 359:1655–1665PubMedCrossRef
81.
go back to reference Scott JA, Ojal J, Ashton L, Muhoro A, Burbidge P, Goldblatt D (2011) Pneumococcal conjugate vaccine given shortly after birth stimulates effective antibody concentrations and primes immunological memory for sustained infant protection. Clin Infect Dis 53:663–670PubMedPubMedCentralCrossRef Scott JA, Ojal J, Ashton L, Muhoro A, Burbidge P, Goldblatt D (2011) Pneumococcal conjugate vaccine given shortly after birth stimulates effective antibody concentrations and primes immunological memory for sustained infant protection. Clin Infect Dis 53:663–670PubMedPubMedCentralCrossRef
82.
go back to reference Clarke E, Kampmann B, Goldblatt D (2016) Maternal and neonatal pneumococcal vaccination—where are we now? Expert Rev Vaccines 15:1305–1317PubMedCrossRef Clarke E, Kampmann B, Goldblatt D (2016) Maternal and neonatal pneumococcal vaccination—where are we now? Expert Rev Vaccines 15:1305–1317PubMedCrossRef
83.
go back to reference Pomat WS, van den Biggelaar AH, Phuanukoonnon S, Francis J, Jacoby P, Siba PM, Alpers MP, Reeder JC, Holt PG, Richmond PC, Lehmann D (2013) Safety and immunogenicity of neonatal pneumococcal conjugate vaccination in Papua New Guinean children: a randomised controlled trial. PLoS One 8:e56698PubMedPubMedCentralCrossRef Pomat WS, van den Biggelaar AH, Phuanukoonnon S, Francis J, Jacoby P, Siba PM, Alpers MP, Reeder JC, Holt PG, Richmond PC, Lehmann D (2013) Safety and immunogenicity of neonatal pneumococcal conjugate vaccination in Papua New Guinean children: a randomised controlled trial. PLoS One 8:e56698PubMedPubMedCentralCrossRef
84.
go back to reference Provenzano RW, Wetterlow LH, Sullivan CL (1965) Immunization and antibody response in the newborn infant. I. Pertussis inoculation within twenty-four hours of birth. N Engl J Med 273:959–965PubMedCrossRef Provenzano RW, Wetterlow LH, Sullivan CL (1965) Immunization and antibody response in the newborn infant. I. Pertussis inoculation within twenty-four hours of birth. N Engl J Med 273:959–965PubMedCrossRef
85.
go back to reference Locht C (2016) Pertussis: acellular, whole-cell, new vaccines, what to choose? Expert Rev Vaccines 15:671–673PubMedCrossRef Locht C (2016) Pertussis: acellular, whole-cell, new vaccines, what to choose? Expert Rev Vaccines 15:671–673PubMedCrossRef
86.
go back to reference Locht C, Mielcarek N (2012) New pertussis vaccination approaches: en route to protect newborns? FEMS Immunol Med Microbiol 66:121–133PubMedCrossRef Locht C, Mielcarek N (2012) New pertussis vaccination approaches: en route to protect newborns? FEMS Immunol Med Microbiol 66:121–133PubMedCrossRef
87.
go back to reference Belloni C, De Silvestri A, Tinelli C, Avanzini MA, Marconi M, Strano F, Rondini G, Chirico G (2003) Immunogenicity of a three-component acellular pertussis vaccine administered at birth. Pediatrics 111:1042–1045PubMedCrossRef Belloni C, De Silvestri A, Tinelli C, Avanzini MA, Marconi M, Strano F, Rondini G, Chirico G (2003) Immunogenicity of a three-component acellular pertussis vaccine administered at birth. Pediatrics 111:1042–1045PubMedCrossRef
88.
go back to reference Wood N, McIntyre P, Marshall H, Roberton D (2010) Acellular pertussis vaccine at birth and one month induces antibody responses by two months of age. Pediatr Infect Dis J 29:209–215PubMedCrossRef Wood N, McIntyre P, Marshall H, Roberton D (2010) Acellular pertussis vaccine at birth and one month induces antibody responses by two months of age. Pediatr Infect Dis J 29:209–215PubMedCrossRef
89.
go back to reference Saso A, Kampmann B (2016) Vaccination against respiratory syncytial virus in pregnancy: a suitable tool to combat global infant morbidity and mortality? Lancet Infect Dis 16:e153–e163PubMedCrossRef Saso A, Kampmann B (2016) Vaccination against respiratory syncytial virus in pregnancy: a suitable tool to combat global infant morbidity and mortality? Lancet Infect Dis 16:e153–e163PubMedCrossRef
90.
go back to reference Saleh E, Eichner B, Clark DW, Gagliano ME, Troutman JM, Harrington L, McNeal M, Clements D (2017) Open-label pilot study to compare the safety and immunogenicity of pentavalent rotavirus vaccine (RV5) administered on an early alternative dosing schedule with those of RV5 administered on the recommended standard schedule. J Pediatric Infect Dis Soc 3. https://doi.org/10.1093/jpids/pix005 Saleh E, Eichner B, Clark DW, Gagliano ME, Troutman JM, Harrington L, McNeal M, Clements D (2017) Open-label pilot study to compare the safety and immunogenicity of pentavalent rotavirus vaccine (RV5) administered on an early alternative dosing schedule with those of RV5 administered on the recommended standard schedule. J Pediatric Infect Dis Soc 3. https://​doi.​org/​10.​1093/​jpids/​pix005
91.
go back to reference Morein B, Blomqvist G, Hu K (2007) Immune responsiveness in the neonatal period. J Comp Pathol 137(Suppl 1):S27–S31PubMedCrossRef Morein B, Blomqvist G, Hu K (2007) Immune responsiveness in the neonatal period. J Comp Pathol 137(Suppl 1):S27–S31PubMedCrossRef
92.
go back to reference Siegrist CA (2003) Mechanisms by which maternal antibodies influence infant vaccine responses: review of hypotheses and definition of main determinants. Vaccine 21:3406–3412PubMedCrossRef Siegrist CA (2003) Mechanisms by which maternal antibodies influence infant vaccine responses: review of hypotheses and definition of main determinants. Vaccine 21:3406–3412PubMedCrossRef
93.
go back to reference Caceres VM, Strebel PM, Sutter RW (2000) Factors determining prevalence of maternal antibody to measles virus throughout infancy: a review. Clin Infect Dis 31:110–119PubMedCrossRef Caceres VM, Strebel PM, Sutter RW (2000) Factors determining prevalence of maternal antibody to measles virus throughout infancy: a review. Clin Infect Dis 31:110–119PubMedCrossRef
94.
go back to reference Pyzik M, Rath T, Lencer WI, Baker K, Blumberg RS (2015) FcRn: the architect behind the immune and nonimmune functions of IgG and albumin. J Immunol 194:4595–4603PubMedPubMedCentralCrossRef Pyzik M, Rath T, Lencer WI, Baker K, Blumberg RS (2015) FcRn: the architect behind the immune and nonimmune functions of IgG and albumin. J Immunol 194:4595–4603PubMedPubMedCentralCrossRef
95.
go back to reference Moon SS, Wang Y, Shane AL, Nguyen T, Ray P, Dennehy P, Baek LJ, Parashar U, Glass RI, Jiang B (2010) Inhibitory effect of breast milk on infectivity of live oral rotavirus vaccines. Pediatr Infect Dis J 29:919–923PubMedPubMedCentralCrossRef Moon SS, Wang Y, Shane AL, Nguyen T, Ray P, Dennehy P, Baek LJ, Parashar U, Glass RI, Jiang B (2010) Inhibitory effect of breast milk on infectivity of live oral rotavirus vaccines. Pediatr Infect Dis J 29:919–923PubMedPubMedCentralCrossRef
96.
go back to reference Dorea JG (2012) Breast-feeding and responses to infant vaccines: constitutional and environmental factors. Am J Perinatol 29:759–775PubMedCrossRef Dorea JG (2012) Breast-feeding and responses to infant vaccines: constitutional and environmental factors. Am J Perinatol 29:759–775PubMedCrossRef
97.
go back to reference Chen MY, Kirkwood CD, Bines J, Cowley D, Pavlic D, Lee KJ, Orsini F, Watts E, Barnes G, Danchin M (2017) Rotavirus specific maternal antibodies and immune response to RV3-BB neonatal rotavirus vaccine in New Zealand. Hum Vaccin Immunother 13:1126–1135PubMedCrossRefPubMedCentral Chen MY, Kirkwood CD, Bines J, Cowley D, Pavlic D, Lee KJ, Orsini F, Watts E, Barnes G, Danchin M (2017) Rotavirus specific maternal antibodies and immune response to RV3-BB neonatal rotavirus vaccine in New Zealand. Hum Vaccin Immunother 13:1126–1135PubMedCrossRefPubMedCentral
98.
go back to reference Ali A, Kazi AM, Cortese MM, Fleming JA, Moon S, Parashar UD, Jiang B, McNeal MM, Steele D, Bhutta Z, Zaidi AK (2015) Impact of withholding breastfeeding at the time of vaccination on the immunogenicity of oral rotavirus vaccine—a randomized trial. PLoS One 10:e0127622PubMedPubMedCentralCrossRef Ali A, Kazi AM, Cortese MM, Fleming JA, Moon S, Parashar UD, Jiang B, McNeal MM, Steele D, Bhutta Z, Zaidi AK (2015) Impact of withholding breastfeeding at the time of vaccination on the immunogenicity of oral rotavirus vaccine—a randomized trial. PLoS One 10:e0127622PubMedPubMedCentralCrossRef
99.
go back to reference Hesseling AC, Johnson LF, Jaspan H, Cotton MF, Whitelaw A, Schaaf HS, Fine PE, Eley BS, Marais BJ, Nuttall J, Beyers N, Godfrey-Faussett P (2009) Disseminated bacille Calmette-Guerin disease in HIV-infected South African infants. Bull World Health Organ 87:505–511PubMedPubMedCentralCrossRef Hesseling AC, Johnson LF, Jaspan H, Cotton MF, Whitelaw A, Schaaf HS, Fine PE, Eley BS, Marais BJ, Nuttall J, Beyers N, Godfrey-Faussett P (2009) Disseminated bacille Calmette-Guerin disease in HIV-infected South African infants. Bull World Health Organ 87:505–511PubMedPubMedCentralCrossRef
100.
go back to reference Hesseling AC, Rabie H, Marais BJ, Manders M, Lips M, Schaaf HS, Gie RP, Cotton MF, van Helden PD, Warren RM, Beyers N (2006) Bacille Calmette-Guerin vaccine-induced disease in HIV-infected and HIV-uninfected children. Clin Infect Dis 42:548–558PubMedCrossRef Hesseling AC, Rabie H, Marais BJ, Manders M, Lips M, Schaaf HS, Gie RP, Cotton MF, van Helden PD, Warren RM, Beyers N (2006) Bacille Calmette-Guerin vaccine-induced disease in HIV-infected and HIV-uninfected children. Clin Infect Dis 42:548–558PubMedCrossRef
101.
go back to reference Dauby N, Goetghebuer T, Kollmann TR, Levy J, Marchant A (2012) Uninfected but not unaffected: chronic maternal infections during pregnancy, fetal immunity, and susceptibility to postnatal infections. Lancet Infect Dis 12:330–340PubMedCrossRef Dauby N, Goetghebuer T, Kollmann TR, Levy J, Marchant A (2012) Uninfected but not unaffected: chronic maternal infections during pregnancy, fetal immunity, and susceptibility to postnatal infections. Lancet Infect Dis 12:330–340PubMedCrossRef
102.
go back to reference Lisciandro JG, van den Biggelaar AH (2010) Neonatal immune function and inflammatory illnesses in later life: lessons to be learnt from the developing world? Clin Exp Allergy 40:1719–1731PubMedCrossRef Lisciandro JG, van den Biggelaar AH (2010) Neonatal immune function and inflammatory illnesses in later life: lessons to be learnt from the developing world? Clin Exp Allergy 40:1719–1731PubMedCrossRef
103.
go back to reference Malhotra I, Ouma J, Wamachi A, Kioko J, Mungai P, Omollo A, Elson L, Koech D, Kazura JW, King CL (1997) In utero exposure to helminth and mycobacterial antigens generates cytokine responses similar to that observed in adults. J Clin Invest 99:1759–1766PubMedPubMedCentralCrossRef Malhotra I, Ouma J, Wamachi A, Kioko J, Mungai P, Omollo A, Elson L, Koech D, Kazura JW, King CL (1997) In utero exposure to helminth and mycobacterial antigens generates cytokine responses similar to that observed in adults. J Clin Invest 99:1759–1766PubMedPubMedCentralCrossRef
104.
go back to reference van Riet E, Hartgers FC, Yazdanbakhsh M (2007) Chronic helminth infections induce immunomodulation: consequences and mechanisms. Immunobiology 212:475–490PubMedCrossRef van Riet E, Hartgers FC, Yazdanbakhsh M (2007) Chronic helminth infections induce immunomodulation: consequences and mechanisms. Immunobiology 212:475–490PubMedCrossRef
105.
go back to reference Malhotra Iea (2010) Antenatal helminth infections are associated with impaired Hib vaccine efficiency in Kenyan infants. In: Keystone symposia: immmunological mechanisms of vaccination. Seattle, WA Malhotra Iea (2010) Antenatal helminth infections are associated with impaired Hib vaccine efficiency in Kenyan infants. In: Keystone symposia: immmunological mechanisms of vaccination. Seattle, WA
106.
go back to reference Malhotra I, Mungai P, Wamachi A, Kioko J, Ouma JH, Kazura JW, King CL (1999) Helminth- and Bacillus Calmette-Guerin-induced immunity in children sensitized in utero to filariasis and schistosomiasis. J Immunol 162:6843–6848PubMed Malhotra I, Mungai P, Wamachi A, Kioko J, Ouma JH, Kazura JW, King CL (1999) Helminth- and Bacillus Calmette-Guerin-induced immunity in children sensitized in utero to filariasis and schistosomiasis. J Immunol 162:6843–6848PubMed
107.
go back to reference Elliott AM, Mawa PA, Webb EL, Nampijja M, Lyadda N, Bukusuba J, Kizza M, Namujju PB, Nabulime J, Ndibazza J, Muwanga M, Whitworth JA (2010) Effects of maternal and infant co-infections, and of maternal immunisation, on the infant response to BCG and tetanus immunisation. Vaccine 29:247–255PubMedPubMedCentralCrossRef Elliott AM, Mawa PA, Webb EL, Nampijja M, Lyadda N, Bukusuba J, Kizza M, Namujju PB, Nabulime J, Ndibazza J, Muwanga M, Whitworth JA (2010) Effects of maternal and infant co-infections, and of maternal immunisation, on the infant response to BCG and tetanus immunisation. Vaccine 29:247–255PubMedPubMedCentralCrossRef
108.
go back to reference Dauby N, Alonso-Vega C, Suarez E, Flores A, Hermann E, Cordova M, Tellez T, Torrico F, Truyens C, Carlier Y (2009) Maternal infection with Trypanosoma cruzi and congenital Chagas disease induce a trend to a type 1 polarization of infant immune responses to vaccines. PLoS Negl Trop Dis 3:e571PubMedPubMedCentralCrossRef Dauby N, Alonso-Vega C, Suarez E, Flores A, Hermann E, Cordova M, Tellez T, Torrico F, Truyens C, Carlier Y (2009) Maternal infection with Trypanosoma cruzi and congenital Chagas disease induce a trend to a type 1 polarization of infant immune responses to vaccines. PLoS Negl Trop Dis 3:e571PubMedPubMedCentralCrossRef
109.
go back to reference Mansoor N, Scriba TJ, de Kock M, Tameris M, Abel B, Keyser A, Little F, Soares A, Gelderbloem S, Mlenjeni S, Denation L, Hawkridge A, Boom WH, Kaplan G, Hussey GD, Hanekom WA (2009) HIV-1 infection in infants severely impairs the immune response induced by Bacille Calmette-Guerin vaccine. J Infect Dis 199:982–990PubMedPubMedCentralCrossRef Mansoor N, Scriba TJ, de Kock M, Tameris M, Abel B, Keyser A, Little F, Soares A, Gelderbloem S, Mlenjeni S, Denation L, Hawkridge A, Boom WH, Kaplan G, Hussey GD, Hanekom WA (2009) HIV-1 infection in infants severely impairs the immune response induced by Bacille Calmette-Guerin vaccine. J Infect Dis 199:982–990PubMedPubMedCentralCrossRef
110.
go back to reference Helfand RF, Witte D, Fowlkes A, Garcia P, Yang C, Fudzulani R, Walls L, Bae S, Strebel P, Broadhead R, Bellini WJ, Cutts F (2008) Evaluation of the immune response to a 2-dose measles vaccination schedule administered at 6 and 9 months of age to HIV-infected and HIV-uninfected children in Malawi. J Infect Dis 198:1457–1465PubMedCrossRef Helfand RF, Witte D, Fowlkes A, Garcia P, Yang C, Fudzulani R, Walls L, Bae S, Strebel P, Broadhead R, Bellini WJ, Cutts F (2008) Evaluation of the immune response to a 2-dose measles vaccination schedule administered at 6 and 9 months of age to HIV-infected and HIV-uninfected children in Malawi. J Infect Dis 198:1457–1465PubMedCrossRef
111.
go back to reference Jones CE, Naidoo S, De Beer C, Esser M, Kampmann B, Hesseling AC (2011) Maternal HIV infection and antibody responses against vaccine-preventable diseases in uninfected infants. JAMA 305:576–584PubMedCrossRef Jones CE, Naidoo S, De Beer C, Esser M, Kampmann B, Hesseling AC (2011) Maternal HIV infection and antibody responses against vaccine-preventable diseases in uninfected infants. JAMA 305:576–584PubMedCrossRef
112.
go back to reference Jones CE, Hesseling AC, Tena-Coki NG, Scriba TJ, Chegou NN, Kidd M, Wilkinson RJ, Kampmann B (2015) The impact of HIV exposure and maternal Mycobacterium tuberculosis infection on infant immune responses to bacille Calmette-Guerin vaccination. AIDS 29:155–165PubMedPubMedCentralCrossRef Jones CE, Hesseling AC, Tena-Coki NG, Scriba TJ, Chegou NN, Kidd M, Wilkinson RJ, Kampmann B (2015) The impact of HIV exposure and maternal Mycobacterium tuberculosis infection on infant immune responses to bacille Calmette-Guerin vaccination. AIDS 29:155–165PubMedPubMedCentralCrossRef
113.
go back to reference Mazzola TN, da Silva MT, Abramczuk BM, Moreno YM, Lima SC, Zorzeto TQ, Passeto AS, Vilela MM (2011) Impaired Bacillus Calmette-Guerin cellular immune response in HIV-exposed, uninfected infants. AIDS 25:2079–2087PubMedCrossRef Mazzola TN, da Silva MT, Abramczuk BM, Moreno YM, Lima SC, Zorzeto TQ, Passeto AS, Vilela MM (2011) Impaired Bacillus Calmette-Guerin cellular immune response in HIV-exposed, uninfected infants. AIDS 25:2079–2087PubMedCrossRef
114.
go back to reference Abramczuk BM, Mazzola TN, Moreno YM, Zorzeto TQ, Quintilio W, Wolf PS, Blotta MH, Morcillo AM, da Silva MT, Dos Santos Vilela MM (2011) Impaired humoral response to vaccines among HIV-exposed uninfected infants. Clin Vaccine Immunol 18:1406–1409PubMedPubMedCentralCrossRef Abramczuk BM, Mazzola TN, Moreno YM, Zorzeto TQ, Quintilio W, Wolf PS, Blotta MH, Morcillo AM, da Silva MT, Dos Santos Vilela MM (2011) Impaired humoral response to vaccines among HIV-exposed uninfected infants. Clin Vaccine Immunol 18:1406–1409PubMedPubMedCentralCrossRef
115.
go back to reference Okoko BJ, Wesumperuma LH, Ota MO, Pinder M, Banya W, Gomez SF, McAdam KP, Hart AC (2001) The influence of placental malaria infection and maternal hypergammaglobulinemia on transplacental transfer of antibodies and IgG subclasses in a rural West African population. J Infect Dis 184:627–632PubMedCrossRef Okoko BJ, Wesumperuma LH, Ota MO, Pinder M, Banya W, Gomez SF, McAdam KP, Hart AC (2001) The influence of placental malaria infection and maternal hypergammaglobulinemia on transplacental transfer of antibodies and IgG subclasses in a rural West African population. J Infect Dis 184:627–632PubMedCrossRef
116.
go back to reference Okoko BJ, Wesuperuma LH, Ota MO, Banya WA, Pinder M, Gomez FS, Osinusi K, Hart AC (2001) Influence of placental malaria infection and maternal hypergammaglobulinaemia on materno-foetal transfer of measles and tetanus antibodies in a rural West African population. J Health Popul Nutr 19:59–65PubMed Okoko BJ, Wesuperuma LH, Ota MO, Banya WA, Pinder M, Gomez FS, Osinusi K, Hart AC (2001) Influence of placental malaria infection and maternal hypergammaglobulinaemia on materno-foetal transfer of measles and tetanus antibodies in a rural West African population. J Health Popul Nutr 19:59–65PubMed
117.
go back to reference Cumberland P, Shulman CE, Maple PA, Bulmer JN, Dorman EK, Kawuondo K, Marsh K, Cutts FT (2007) Maternal HIV infection and placental malaria reduce transplacental antibody transfer and tetanus antibody levels in newborns in Kenya. J Infect Dis 196:550–557PubMedCrossRef Cumberland P, Shulman CE, Maple PA, Bulmer JN, Dorman EK, Kawuondo K, Marsh K, Cutts FT (2007) Maternal HIV infection and placental malaria reduce transplacental antibody transfer and tetanus antibody levels in newborns in Kenya. J Infect Dis 196:550–557PubMedCrossRef
118.
go back to reference Ned RM, Price AE, Crawford SB, Ayisi JG, van Eijk AM, Otieno JA, Nahlen BL, Steketee RW, Slutsker L, Shi YP, Lanar DE, Udhayakumar V (2008) Effect of placental malaria and HIV infection on the antibody responses to Plasmodium falciparum in infants. J Infect Dis 198:1609–1619PubMedCrossRef Ned RM, Price AE, Crawford SB, Ayisi JG, van Eijk AM, Otieno JA, Nahlen BL, Steketee RW, Slutsker L, Shi YP, Lanar DE, Udhayakumar V (2008) Effect of placental malaria and HIV infection on the antibody responses to Plasmodium falciparum in infants. J Infect Dis 198:1609–1619PubMedCrossRef
119.
go back to reference Newport MJ, Goetghebuer T, Weiss HA, Whittle H, Siegrist CA, Marchant A (2004) Genetic regulation of immune responses to vaccines in early life. Genes Immun 5:122–129PubMedCrossRef Newport MJ, Goetghebuer T, Weiss HA, Whittle H, Siegrist CA, Marchant A (2004) Genetic regulation of immune responses to vaccines in early life. Genes Immun 5:122–129PubMedCrossRef
120.
go back to reference Marchant A, Pihlgren M, Goetghebuer T, Weiss HA, Ota MO, Schlegel-Hauter SE, Whittle H, Lambert PH, Newport MJ, Siegrist CA (2006) Predominant influence of environmental determinants on the persistence and avidity maturation of antibody responses to vaccines in infants. J Infect Dis 193:1598–1605PubMedCrossRef Marchant A, Pihlgren M, Goetghebuer T, Weiss HA, Ota MO, Schlegel-Hauter SE, Whittle H, Lambert PH, Newport MJ, Siegrist CA (2006) Predominant influence of environmental determinants on the persistence and avidity maturation of antibody responses to vaccines in infants. J Infect Dis 193:1598–1605PubMedCrossRef
121.
go back to reference Mentzer AJ, O'Connor D, Pollard AJ, Hill AV (2015) Searching for the human genetic factors standing in the way of universally effective vaccines. Philos Trans R Soc Lond Ser B Biol Sci 19;370(1671) Mentzer AJ, O'Connor D, Pollard AJ, Hill AV (2015) Searching for the human genetic factors standing in the way of universally effective vaccines. Philos Trans R Soc Lond Ser B Biol Sci 19;370(1671)
124.
go back to reference Blondheim O, Bader D, Abend M, Peniakov M, Reich D, Potesman I, Handsher R, Gidoni I, Linder N (1998) Immunogenicity of hepatitis B vaccine in preterm infants. Arch Dis Child Fetal Neonatal Ed 79:F206–F208PubMedPubMedCentralCrossRef Blondheim O, Bader D, Abend M, Peniakov M, Reich D, Potesman I, Handsher R, Gidoni I, Linder N (1998) Immunogenicity of hepatitis B vaccine in preterm infants. Arch Dis Child Fetal Neonatal Ed 79:F206–F208PubMedPubMedCentralCrossRef
125.
go back to reference Linder N, Vishne TH, Levin E, Handsher R, Fink-Kremer I, Waldman D, Levine A, Ashkenazi S, Sirota L (2002) Hepatitis B vaccination: long-term follow-up of the immune response of preterm infants and comparison of two vaccination protocols. Infection 30:136–139PubMedCrossRef Linder N, Vishne TH, Levin E, Handsher R, Fink-Kremer I, Waldman D, Levine A, Ashkenazi S, Sirota L (2002) Hepatitis B vaccination: long-term follow-up of the immune response of preterm infants and comparison of two vaccination protocols. Infection 30:136–139PubMedCrossRef
126.
go back to reference Marciano BE, Huang CY, Joshi G, Rezaei N, Carvalho BC, Allwood Z, Ikinciogullari A, Reda SM, Gennery A, Thon V, Espinosa-Rosales F, Al-Herz W, Porras O, Shcherbina A, Szaflarska A, Kilic S, Franco JL, Gomez Raccio AC, Roxo P Jr, Esteves I, Galal N, Grumach AS, Al-Tamemi S, Yildiran A, Orellana JC, Yamada M, Morio T, Liberatore D, Ohtsuka Y, Lau YL, Nishikomori R, Torres-Lozano C, Mazzucchelli JT, Vilela MM, Tavares FS, Cunha L, Pinto JA, Espinosa-Padilla SE, Hernandez-Nieto L, Elfeky RA, Ariga T, Toshio H, Dogu F, Cipe F, Formankova R, Nunez-Nunez ME, Bezrodnik L, Marques JG, Pereira MI, Listello V, Slatter MA, Nademi Z, Kowalczyk D, Fleisher TA, Davies G, Neven B, Rosenzweig SD (2014) BCG vaccination in patients with severe combined immunodeficiency: complications, risks, and vaccination policies. J Allergy Clin Immunol 133:1134–1141PubMedPubMedCentralCrossRef Marciano BE, Huang CY, Joshi G, Rezaei N, Carvalho BC, Allwood Z, Ikinciogullari A, Reda SM, Gennery A, Thon V, Espinosa-Rosales F, Al-Herz W, Porras O, Shcherbina A, Szaflarska A, Kilic S, Franco JL, Gomez Raccio AC, Roxo P Jr, Esteves I, Galal N, Grumach AS, Al-Tamemi S, Yildiran A, Orellana JC, Yamada M, Morio T, Liberatore D, Ohtsuka Y, Lau YL, Nishikomori R, Torres-Lozano C, Mazzucchelli JT, Vilela MM, Tavares FS, Cunha L, Pinto JA, Espinosa-Padilla SE, Hernandez-Nieto L, Elfeky RA, Ariga T, Toshio H, Dogu F, Cipe F, Formankova R, Nunez-Nunez ME, Bezrodnik L, Marques JG, Pereira MI, Listello V, Slatter MA, Nademi Z, Kowalczyk D, Fleisher TA, Davies G, Neven B, Rosenzweig SD (2014) BCG vaccination in patients with severe combined immunodeficiency: complications, risks, and vaccination policies. J Allergy Clin Immunol 133:1134–1141PubMedPubMedCentralCrossRef
127.
go back to reference Hartgers FC, Obeng BB, Voskamp A, Larbi IA, Amoah AS, Luty AJ, Boakye D, Yazdanbakhsh M (2008) Enhanced Toll-like receptor responsiveness associated with mitogen-activated protein kinase activation in Plasmodium falciparum-infected children. Infect Immun 76:5149–5157PubMedPubMedCentralCrossRef Hartgers FC, Obeng BB, Voskamp A, Larbi IA, Amoah AS, Luty AJ, Boakye D, Yazdanbakhsh M (2008) Enhanced Toll-like receptor responsiveness associated with mitogen-activated protein kinase activation in Plasmodium falciparum-infected children. Infect Immun 76:5149–5157PubMedPubMedCentralCrossRef
128.
go back to reference Lalor MK, Ben-Smith A, Gorak-Stolinska P, Weir RE, Floyd S, Blitz R, Mvula H, Newport MJ, Branson K, McGrath N, Crampin AC, Fine PE, Dockrell HM (2009) Population differences in immune responses to Bacille Calmette-Guerin vaccination in infancy. J Infect Dis 199:795–800PubMedPubMedCentralCrossRef Lalor MK, Ben-Smith A, Gorak-Stolinska P, Weir RE, Floyd S, Blitz R, Mvula H, Newport MJ, Branson K, McGrath N, Crampin AC, Fine PE, Dockrell HM (2009) Population differences in immune responses to Bacille Calmette-Guerin vaccination in infancy. J Infect Dis 199:795–800PubMedPubMedCentralCrossRef
129.
go back to reference Hur YG, Gorak-Stolinska P, Lalor MK, Mvula H, Floyd S, Raynes J, Ben-Smith A, Fitchett JR, Flanagan KL, Burl S, Ota MO, Crampin AC, Smith SG, Dockrell HM (2014) Factors affecting immunogenicity of BCG in infants, a study in Malawi, The Gambia and the UK. BMC Infect Dis 14:184PubMedPubMedCentralCrossRef Hur YG, Gorak-Stolinska P, Lalor MK, Mvula H, Floyd S, Raynes J, Ben-Smith A, Fitchett JR, Flanagan KL, Burl S, Ota MO, Crampin AC, Smith SG, Dockrell HM (2014) Factors affecting immunogenicity of BCG in infants, a study in Malawi, The Gambia and the UK. BMC Infect Dis 14:184PubMedPubMedCentralCrossRef
130.
132.
go back to reference Oh JZ, Ravindran R, Chassaing B, Carvalho FA, Maddur MS, Bower M, Hakimpour P, Gill KP, Nakaya HI, Yarovinsky F, Sartor RB, Gewirtz AT, Pulendran B (2014) TLR5-mediated sensing of gut microbiota is necessary for antibody responses to seasonal influenza vaccination. Immunity 41:478–492PubMedPubMedCentralCrossRef Oh JZ, Ravindran R, Chassaing B, Carvalho FA, Maddur MS, Bower M, Hakimpour P, Gill KP, Nakaya HI, Yarovinsky F, Sartor RB, Gewirtz AT, Pulendran B (2014) TLR5-mediated sensing of gut microbiota is necessary for antibody responses to seasonal influenza vaccination. Immunity 41:478–492PubMedPubMedCentralCrossRef
133.
go back to reference Huda MN, Lewis Z, Kalanetra KM, Rashid M, Ahmad SM, Raqib R, Qadri F, Underwood MA, Mills DA, Stephensen CB (2014) Stool microbiota and vaccine responses of infants. Pediatrics 134:e362–e372PubMedPubMedCentralCrossRef Huda MN, Lewis Z, Kalanetra KM, Rashid M, Ahmad SM, Raqib R, Qadri F, Underwood MA, Mills DA, Stephensen CB (2014) Stool microbiota and vaccine responses of infants. Pediatrics 134:e362–e372PubMedPubMedCentralCrossRef
134.
go back to reference Chappuis G (1998) Neonatal immunity and immunisation in early age: lessons from veterinary medicine. Vaccine 16:1468–1472PubMedCrossRef Chappuis G (1998) Neonatal immunity and immunisation in early age: lessons from veterinary medicine. Vaccine 16:1468–1472PubMedCrossRef
135.
go back to reference de Brito CA, Goldoni AL, Sato MN (2009) Immune adjuvants in early life: targeting the innate immune system to overcome impaired adaptive response. Immunotherapy 1:883–895PubMedCrossRef de Brito CA, Goldoni AL, Sato MN (2009) Immune adjuvants in early life: targeting the innate immune system to overcome impaired adaptive response. Immunotherapy 1:883–895PubMedCrossRef
138.
go back to reference Goriely S, Van Lint C, Dadkhah R, Libin M, De Wit D, Demonte D, Willems F, Goldman M (2004) A defect in nucleosome remodeling prevents IL-12(p35) gene transcription in neonatal dendritic cells. J Exp Med 199:1011–1016PubMedPubMedCentralCrossRef Goriely S, Van Lint C, Dadkhah R, Libin M, De Wit D, Demonte D, Willems F, Goldman M (2004) A defect in nucleosome remodeling prevents IL-12(p35) gene transcription in neonatal dendritic cells. J Exp Med 199:1011–1016PubMedPubMedCentralCrossRef
139.
go back to reference Arulanandam BP, Mittler JN, Lee WT, O'Toole M, Metzger DW (2000) Neonatal administration of IL-12 enhances the protective efficacy of antiviral vaccines. J Immunol 164:3698–3704PubMedCrossRef Arulanandam BP, Mittler JN, Lee WT, O'Toole M, Metzger DW (2000) Neonatal administration of IL-12 enhances the protective efficacy of antiviral vaccines. J Immunol 164:3698–3704PubMedCrossRef
140.
go back to reference Krumbiegel D, Zepp F, Meyer CU (2007) Combined Toll-like receptor agonists synergistically increase production of inflammatory cytokines in human neonatal dendritic cells. Hum Immunol 68:813–822PubMedCrossRef Krumbiegel D, Zepp F, Meyer CU (2007) Combined Toll-like receptor agonists synergistically increase production of inflammatory cytokines in human neonatal dendritic cells. Hum Immunol 68:813–822PubMedCrossRef
141.
go back to reference Philbin VJ, Dowling DJ, Gallington LC, Cortes G, Tan Z, Suter EE, Chi KW, Shuckett A, Stoler-Barak L, Tomai M, Miller RL, Mansfield K, Levy O (2012) Imidazoquinoline Toll-like receptor 8 agonists activate human newborn monocytes and dendritic cells through adenosine-refractory and caspase-1-dependent pathways. J Allergy Clin Immunol 130:195–204.e9PubMedPubMedCentralCrossRef Philbin VJ, Dowling DJ, Gallington LC, Cortes G, Tan Z, Suter EE, Chi KW, Shuckett A, Stoler-Barak L, Tomai M, Miller RL, Mansfield K, Levy O (2012) Imidazoquinoline Toll-like receptor 8 agonists activate human newborn monocytes and dendritic cells through adenosine-refractory and caspase-1-dependent pathways. J Allergy Clin Immunol 130:195–204.e9PubMedPubMedCentralCrossRef
142.
go back to reference Gracia A, Polewicz M, Halperin SA, Hancock RE, Potter AA, Babiuk LA, Gerdts V (2011) Antibody responses in adult and neonatal BALB/c mice to immunization with novel Bordetella pertussis vaccine formulations. Vaccine 29:1595–1604PubMedCrossRef Gracia A, Polewicz M, Halperin SA, Hancock RE, Potter AA, Babiuk LA, Gerdts V (2011) Antibody responses in adult and neonatal BALB/c mice to immunization with novel Bordetella pertussis vaccine formulations. Vaccine 29:1595–1604PubMedCrossRef
143.
go back to reference Levy O, Suter EE, Miller RL, Wessels MR (2006) Unique efficacy of Toll-like receptor 8 agonists in activating human neonatal antigen-presenting cells. Blood 108:1284–1290PubMedPubMedCentralCrossRef Levy O, Suter EE, Miller RL, Wessels MR (2006) Unique efficacy of Toll-like receptor 8 agonists in activating human neonatal antigen-presenting cells. Blood 108:1284–1290PubMedPubMedCentralCrossRef
144.
go back to reference Peng G, Guo Z, Kiniwa Y, Voo KS, Peng W, Fu T, Wang DY, Li Y, Wang HY, Wang RF (2005) Toll-like receptor 8-mediated reversal of CD4+ regulatory T cell function. Science 309:1380–1384PubMedCrossRef Peng G, Guo Z, Kiniwa Y, Voo KS, Peng W, Fu T, Wang DY, Li Y, Wang HY, Wang RF (2005) Toll-like receptor 8-mediated reversal of CD4+ regulatory T cell function. Science 309:1380–1384PubMedCrossRef
145.
go back to reference Dowling DJ, Scott EA, Scheid A, Bergelson I, Joshi S, Pietrasanta C, Brightman S, Sanchez-Schmitz G, Van Haren SD, Ninkovic J, Kats D, Guiducci C, de Titta A, Bonner DK, Hirosue S, Swartz MA, Hubbell JA, Levy O (2017) Toll-like receptor 8 agonist nanoparticles mimic immunomodulating effects of the live BCG vaccine and enhance neonatal innate and adaptive immune responses. J Allergy Clin Immunol pii: S0091-6749(17)30242-7. https://doi.org/10.1016/j.jaci.2016.12.985 Dowling DJ, Scott EA, Scheid A, Bergelson I, Joshi S, Pietrasanta C, Brightman S, Sanchez-Schmitz G, Van Haren SD, Ninkovic J, Kats D, Guiducci C, de Titta A, Bonner DK, Hirosue S, Swartz MA, Hubbell JA, Levy O (2017) Toll-like receptor 8 agonist nanoparticles mimic immunomodulating effects of the live BCG vaccine and enhance neonatal innate and adaptive immune responses. J Allergy Clin Immunol pii: S0091-6749(17)30242-7. https://​doi.​org/​10.​1016/​j.​jaci.​2016.​12.​985
146.
go back to reference Noh Y, Shim BS, Cheon IS, Rho S, Kim HJ, Choi Y, Kang CY, Chang J, Song MK, Kim JO (2013) Neonatal immunization with respiratory syncytial virus glycoprotein fragment induces protective immunity in the presence of maternal antibodies in mice. Viral Immunol 26:268–276PubMedPubMedCentralCrossRef Noh Y, Shim BS, Cheon IS, Rho S, Kim HJ, Choi Y, Kang CY, Chang J, Song MK, Kim JO (2013) Neonatal immunization with respiratory syncytial virus glycoprotein fragment induces protective immunity in the presence of maternal antibodies in mice. Viral Immunol 26:268–276PubMedPubMedCentralCrossRef
147.
go back to reference Liang ZZ, Sherrid AM, Wallecha A, Kollmann TR (2014) Listeria monocytogenes: a promising vehicle for neonatal vaccination. Hum Vaccin Immunother 10:1036–1046PubMedPubMedCentralCrossRef Liang ZZ, Sherrid AM, Wallecha A, Kollmann TR (2014) Listeria monocytogenes: a promising vehicle for neonatal vaccination. Hum Vaccin Immunother 10:1036–1046PubMedPubMedCentralCrossRef
148.
go back to reference Dai G, Rady HF, Huang W, Shellito JE, Mason C, Ramsay AJ (2016) Gene-based neonatal immune priming potentiates a mucosal adenoviral vaccine encoding mycobacterial Ag85B. Vaccine 34:6267–6275PubMedCrossRefPubMedCentral Dai G, Rady HF, Huang W, Shellito JE, Mason C, Ramsay AJ (2016) Gene-based neonatal immune priming potentiates a mucosal adenoviral vaccine encoding mycobacterial Ag85B. Vaccine 34:6267–6275PubMedCrossRefPubMedCentral
149.
go back to reference Sedegah M, Hoffman SL (2006) Immunological responses of neonates and infants to DNA vaccines. Methods Mol Med 127:239–251PubMed Sedegah M, Hoffman SL (2006) Immunological responses of neonates and infants to DNA vaccines. Methods Mol Med 127:239–251PubMed
Metadata
Title
Vaccine responses in newborns
Authors
Anja Saso
Beate Kampmann
Publication date
01-11-2017
Publisher
Springer Berlin Heidelberg
Published in
Seminars in Immunopathology / Issue 6/2017
Print ISSN: 1863-2297
Electronic ISSN: 1863-2300
DOI
https://doi.org/10.1007/s00281-017-0654-9

Other articles of this Issue 6/2017

Seminars in Immunopathology 6/2017 Go to the issue