Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Dec 1.
Published in final edited form as: Curr Opin Physiol. 2018 Mar 29;6:16–20. doi: 10.1016/j.cophys.2018.03.010

The evolution of greater humoral immunity in females than males: implications for vaccine efficacy

Ashley L Fink 1, Sabra L Klein 1
PMCID: PMC6181235  NIHMSID: NIHMS958084  PMID: 30320243

Abstract

Males and females differ in their effector and memory immune responses to foreign and self-antigens. The difference in antibody responses (i.e., humoral immunity), in particular, is one of the most well conserved sex differences in immunology. Certain sex differences in humoral immunity are present throughout life, whereas others are only apparent after puberty and prior to reproductive senescence, suggesting that both genes and hormones are involved. Importantly, these sex-based differences in humoral immunity contribute to variation in the responses to vaccines and may explain some disparities in vaccine efficacy between the sexes. Elevated humoral immunity in females compared with males is phylogenetically well conserved, suggesting an adaptive advantage of elevated antibody for reproductive success, including for the transfer of protective antibodies to offspring.

Keywords: antibody, B lymphocyte, estrogen, immunization, influenza, X chromosome


The term ‘sex’ is a biological construct that defines males and females by the basic organization of chromosomes, reproductive organs, and circulating sex steroid hormone concentrations. There is a growing appreciation that sex is a biological variable that can impact immune responses through genetic and hormonal regulation of immune cells [1]. One of the most well conserved immunological differences between the sexes is in antibody responses to foreign antigens [2]. Generally, among adults of reproductive ages, females have greater antibody responses than males, higher basal immunoglobulin (Ig) levels, and higher B cell numbers [35]. Adult females also have higher antibody responses to diverse vaccines against, influenza, hepatitis B, yellow fever, rabies, herpes, and smallpox viruses [2]. Higher antibody responses to vaccine antigens imply that the efficacy of vaccines should be greater in females than males, which has not been adequately analyzed in epidemiological studies. The phylogenetic and ontogenetic development of sex differences in humoral immunity illustrate that this conserved immunological difference between the sexes has functional significance, including for vaccine efficacy, and is profoundly affected by both genes and hormones.

Ontogeny of sex differences in humoral immunity

The effects of age on the humoral immune response to vaccination are well documented, yet very few studies consider both age and sex as biological variables. Sex differences are most profound in individuals after sexual maturation, and the underlying mechanisms for these differences have been attributed to both hormonal and genetic effects on the immune system. However, the interplay between age, specifically prior to sexual maturation and following reproductive senescence, and the differential outcomes between the sexes following immune stimulation requires greater consideration.

Pre-puberty

The humoral immune response to vaccinations in children prior to puberty is functionally and quantitatively different compared with that of older children and adults. After birth, infants are exposed to various foreign antigens and their immune system functions to reach a balance between immunotolerance and immunoreactivity. The CD4+ T cell responses in infants are skewed toward anti-inflammatory T-helper (Th)-2 and T-regulatory (Treg) responses, correlating with a lower capacity of antigen presenting cells to produce Th1 polarizing cytokines [6]. The B cell arm of the adaptive immune response also differs between young children and adults. Naïve B cells in infants have lower expression of the co-receptors CD80, CD86, CD40, and HLA-peptide, that interact with T cell ligands during T cell-dependent B cell activation and production of antibody [7,8]. Infants also have lower levels of CD21, the receptor for the C3 complement activation component C3d, which also dampens T cell-dependent B cell activation and impairs germinal center activation [9]. Furthermore, early-life B cells show decreased somatic hypermutation, and bone marrow stromal cells fail to support long-term plasmablast survival and differentiation into plasma cells [10,11]. As a result, infant antibody responses are generally delayed, have lower affinity for the target antigen, and decline more rapidly than those elicited in adults.

There are few reports of sex differences in the humoral immune response in infants and children prior to puberty. Numbers of B cells and levels of IgG and IgM are equivalent between the sexes; whereas infant males are reported to have higher IgA and IgE than females [1]. Sex differences in the B-cell activating factor (BAFF) are present in cord blood from infants, with girls having greater levels of BAFF at birth than boys [12]. The sexes differ in immature/naïve CD5+ B cells during the first three years of life, with boys having greater proportions of CD5+ B cells at birth to 5 days, 4 months, and 36 months of age relative to girls [12]. The observed combination of lower levels of BAFF and higher proportions of CD5+ B cells in boys as compared with girls suggests that sex differences in the humoral immune response may be present at birth, with boys having a more immature/naïve immune system, in general.

Reproductive Ages

Adult females of diverse species tend to have higher antibody responses than males [1]. In adult nonhuman primates, following vaccination against simian immunodeficiency virus (SIV), females show increased local IgA antibody titers, numbers of memory B cells, and numbers of plasma cells as compared to males [13,14]. SIV-vaccinated females also have elevated levels of virus-specific IgG1, IgG2, and IgG3 antibodies as compared to males, with anti-SIV IgG3 antibody levels correlating with antibody mediated cytotoxicity and phagocytic activity, suggesting that the quality of the antibody response is greater in females than males [13]. Studies in mice further illustrate that following vaccination or challenge, females develop higher systemic and mucosal antibody responses than males [15], suggesting that mice are a representative model to study sex differences in humoral immunity.

In humans, global analysis of B cell gene expression signatures reveals that the majority of genes differentially expressed between the sexes are significantly upregulated in B cells from adult females compared with males [16]. Whether these differences in B cell activity reflect intrinsic, genetic differences in B cells between the sexes, or environmental factors, such as hormones, that regulate the functioning of B cells requires greater consideration. The observation that estrogen induces somatic hypermutation and class switch recombination in B cells via the upregulation of activation-induced deaminase, which contains an estrogen response element [17], illustrates that the hormonal milieu can have profound effects on the regulation of B cell activity and antibody production.

Reproductive Senescence

The period of reproductive senescence overlaps with a profound remodeling and decline of the immune system [8]. Changes in the humoral immune response during aging are associated with an increased susceptibility to infectious diseases and increased morbidity and mortality in older adults [18], but the extent to which the age-related decline in the humoral immune response differs between the sexes is poorly understood. Males and females appear to experience the same immune related changes with age, but these changes occur earlier in males as compared with females. For example, males experience a more dramatic decrease in total numbers of T and B cells and larger increases in senescent CD8+ T effector memory cells that re-express the naïve marker CD45 RA (TEMRA) as compared with females [1923]. In addition, a greater proportion of aged males than females demonstrate an inverted CD4:CD8 T cell ratio, an age-related phenotype that is also associated with decreased levels of CD19+ B cells and CD8+CD28- senescent T cells [20]. In contrast, aged females have greater numbers of age associated B cells (ABC) than young females and males of all ages [24,25]. This phenomenon has also been reported in animal models where investigators determined that the mechanism of ABC induction relies on the presence of toll-like receptor 7 (TLR-7) signaling [25,26]. The TLR-7 gene is located on the X chromosome, has biallelic expression in B cells, and may explain how numbers of ABCs are greater in females than males [25,27].

The hormonal milieu changes dramatically with age, in which ovarian function in women and the testicular production of sex steroids in men both decline over time, but at much different rates [28]. In females, the hormonal changes during menopause, specifically declining levels of estrogen and progesterone, occur rapidly [29]; whereas in males, the reduction in testosterone is much more gradual [28,29]. Changes in sex steroid concentrations as well as sex steroid receptor signaling likely contribute to the age and sex-specific changes and dysfunction in the humoral immune response [30]. The hormonal changes associated with menopause (surgical or naturally occurring) in females are directly associated with increased concentrations of proinflammatory cytokines (e.g., TNF-α, IL-1β, and IL-6) and decreased numbers of T and B cells [3133]. Furthermore, the use of hormone replacement therapy in post-menopausal women is known to increase numbers of circulating B cells and reduce baseline concentrations of proinflammatory cytokines as compared to post-menopausal women who are not receiving hormone replacement therapy [32,34]. Males experience a similar decline in numbers of circulating B cells with age but whether or not this is directly a result of decreased testosterone levels has not been determined.

Functional significance of sex differences in humoral immunity for vaccines

Increased susceptibility to infections in infants has led to the development of many vaccines that provide protection prior to exposure. Sex differences have been reported in the antibody responses to many childhood vaccines, however these data are conflicting and limited by the number of studies that report the sex of the individuals and/or partition their data by sex. Antibody responses to diphtheria, pertussis, hepatitis A, hepatitis B, pneumococcal, rabies, human papilloma virus (HPV), and rubella vaccines, as well as to the candidate malaria vaccine RTS,S/ASO2 are higher in girls as compared with boys [1,3539]. Sex differences have also been reported in the antibody responses to the measles vaccine where girls have higher titers as compared to boys, however these data are inconsistent and depend on the vaccine formulation [40]. In contrast, no sex differences have been observed in antibody responses to infant whole cell and acellular pertussis, infant diphtheria and tetanus, meningococcus A and C, or varicella vaccines in children [36,39,41].

Sex differences in the immune responses to vaccines in adults of reproductive ages are well documented, in which females consistently generate greater antibody responses to both live and inactivated vaccines (e.g. influenza, hepatitis B, yellow fever, rabies, herpes, and smallpox) as compared to males [1]. Sex difference in the antibody responses to influenza vaccines are particularly well studied where elevated neutralizing antibody titers are observed against H1N1, H3N2, and influenza B viruses. In adults aged 18–49, receipt of either a half dose or full dose of the seasonal trivalent inactivated vaccine results in hemagglutination inhibition antibody titers that are twice as high in females as compared with males [42]. In response to the hepatitis B vaccine males have an increased nonresponse rate and a more rapid decline in antibody titers with age as compared with females [43].

In aged individuals, sex differences in antibody responses to vaccines are less consistent and depend on the vaccine antigen. Among individual 65+ years of age, hemagglutination inhibition antibody titers to both the standard and high dose seasonal trivalent inactivated influenza vaccine are significantly higher in females as compared to males [44]. In contrast, aged males have higher antibody responses to the tetanus diphtheria and pertussis vaccines as well as the 7-valent and 23-valent pneumococcal vaccines [4549]. The variability observed in the sex differences in responses to vaccines in the elderly are likely a reflection of both biological and behavioral differences between the sexes. Furthermore, the evidence for sex-specific differences in both the rate at which the immune system ages and the differential changes in immune cell populations between the sexes during aging, highlights a need to better consider how age and sex impact the immune responses to vaccines administered to the elderly.

A very limited number of studies have evaluated the impact of sex steroids on immune responses to vaccines. Estradiol, at physiological concentrations, can stimulate antibody production by B cells [50], including antibody responses to an inactivated influenza vaccine administered in mice [51] suggesting one possible mechanism mediating higher antibody production in females, at least prior to reproductive senescence. In humans, reduced neutralizing antibody responses to influenza vaccination are correlated with higher serum testosterone concentrations [5]. Elevated testosterone concentrations in males are also associated with greater lipid metabolism, suggesting that the immunosuppressive role of testosterone and the reduced antibody responses to vaccines in males may be mediated by the expression of genes involved in lipid metabolism that are associated with the suppression of inflammatory responses [5].

Phylogeny of sex differences in humoral immunity

Sex differences in humoral immune responses have evolved in diverse species. In birds, for example, females exhibit higher antibody and cell-mediated immune responses to immune challenges and these effects are often most pronounced during the mating season when male testosterone concentrations are highest [52,53]. Mounting adaptive immune responses that are necessary for clearance of microbes requires metabolic resources that might otherwise be used for other biological processes, such as growth, maintenance of secondary sex characteristics, and reproduction. Trade-offs likely exist for life strategies that impact survival and reproduction [54]. Several theories posit that increased microbial loads and reduced immune function among males are an adverse side effect of positive selection for other traits or characteristics, such as aggressive behavior and the development of male secondary sex characteristics, that increase reproductive success and survival [54].

Although considerable attention has been paid to the evolution of reduced immunity in males, less attention has been paid to the evolution of greater immunity in females. When offspring are young (i.e., during the neonatal period), their immune system has not matured (see above), and protection against infection is primarily mediated by passive immunity from maternal-derived IgG antibodies against microbes. In humans, a majority of maternal antibodies are transferred into fetal circulation, prior to birth, through the placenta [55]. In some mammals, transfer of maternal antibodies can also occur through colostrum immediately following birth and in milk for a long duration after birth [55]. Regardless of species, elevated production of antibodies caused by either vaccination or infection of females enhances the transfer of antibodies to the fetus or neonate to protect them during a critical period of infectious disease susceptibility. We postulate that natural selection favors increased antibody production in females compared with males of reproductive ages because transfer of maternal antibodies from mother to young increases reproductive success by minimizing the detrimental effects of infection on offspring survival.

Concluding remarks

Males and females are biologically different, which impacts adaptive immune responses to diverse antigens, including vaccine antigens. The mechanisms mediating these differences, both hormonal and genetic factors, can alter humoral immune responses to vaccination and may result in sex-specific difference in vaccine efficacy across the life course. Consideration of sex differences in the design of vaccines, including those that protect against influenza, may increase vaccine efficacy. The evolutionary advantage of elevated humoral immunity in females might be the ability to transmit antibodies generated against specific microbes across the placenta and in milk to protect offspring early in life when they are most susceptible to infectious diseases.

Highlights.

  • Among vertebrates, females develop higher antibody responses to diverse antigens than males.

  • Sex differences in antibody responses are reported across the lifespan, but are greatest during reproductive ages (i.e., after puberty and prior to reproductive senescence).

  • Sex differences in humoral immunity likely involve both genetic and hormonal influences that differentially impact B cell functioning in males and females.

  • Higher B cell activity, including antibody production and activity of memory B cells, in females might improve vaccine efficacy in females compared with males.

  • The evolutionary advantage of elevated humoral immunity in females might be the ability to transmit antibodies generated against specific microbes across the placenta and in milk to protect offspring early in life when they are most susceptible to infectious diseases.

Acknowledgments

The writing of this review was supported by the NIH/NIAID Center of Excellence in Influenza Research and Surveillance contract HHS N272201400007C.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of interest statement

Nothing to declare

References

  • **1.Klein SL, Flanagan KL. Sex differences in immune responses. Nat Rev Immunol. 2016;16:626–638. doi: 10.1038/nri.2016.90. A comprehensive review of sex differences in innate and adaptive immune responses with detailed consideration of the mechanisms and functional significance of these differences. [DOI] [PubMed] [Google Scholar]
  • **2.Flanagan KL, Fink AL, Plebanski M, Klein SL. Sex and Gender Differences in the Outcomes of Vaccination over the Life Course. Annu Rev Cell Dev Biol. 2017;33:577–599. doi: 10.1146/annurev-cellbio-100616-060718. A detailed review of the state of knowledge about sex differences in immune responses to vaccines prior to puberty, after puberty and into adulthood, and after reproductive scenescence. [DOI] [PubMed] [Google Scholar]
  • 3.Abdullah M, Chai PS, Chong MY, Tohit ER, Ramasamy R, Pei CP, Vidyadaran S. Gender effect on in vitro lymphocyte subset levels of healthy individuals. Cell Immunol. 2012;272:214–219. doi: 10.1016/j.cellimm.2011.10.009. [DOI] [PubMed] [Google Scholar]
  • 4.Teixeira D, Longo-Maugeri IM, Santos JL, Duarte YA, Lebrao ML, Bueno V. Evaluation of lymphocyte levels in a random sample of 218 elderly individuals from Sao Paulo city. Rev Bras Hematol Hemoter. 2011;33:367–371. doi: 10.5581/1516-8484.20110100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *5.Furman D, Hejblum BP, Simon N, Jojic V, Dekker CL, Thiebaut R, Tibshirani RJ, Davis MM. Systems analysis of sex differences reveals an immunosuppressive role for testosterone in the response to influenza vaccination. Proc Natl Acad Sci U S A. 2014;111:869–874. doi: 10.1073/pnas.1321060111. First consideration of the association between sex steroids and immune responses to vaccines. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kollmann TR, Kampmann B, Mazmanian SK, Marchant A, Levy O. Protecting the Newborn and Young Infant from Infectious Diseases: Lessons from Immune Ontogeny. Immunity. 2017;46:350–363. doi: 10.1016/j.immuni.2017.03.009. [DOI] [PubMed] [Google Scholar]
  • 7.Kaur K, Chowdhury S, Greenspan NS, Schreiber JR. Decreased expression of tumor necrosis factor family receptors involved in humoral immune responses in preterm neonates. Blood. 2007;110:2948–2954. doi: 10.1182/blood-2007-01-069245. [DOI] [PubMed] [Google Scholar]
  • 8.Simon AK, Hollander GA, McMichael A. Evolution of the immune system in humans from infancy to old age. Proc Biol Sci. 2015;282:20143085. doi: 10.1098/rspb.2014.3085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Marchbank KJ, Kulik L, Gipson MG, Morgan BP, Holers VM. Expression of human complement receptor type 2 (CD21) in mice during early B cell development results in a reduction in mature B cells and hypogammaglobulinemia. J Immunol. 2002;169:3526–3535. doi: 10.4049/jimmunol.169.7.3526. [DOI] [PubMed] [Google Scholar]
  • 10.Pihlgren M, Friedli M, Tougne C, Rochat AF, Lambert PH, Siegrist CA. Reduced ability of neonatal and early-life bone marrow stromal cells to support plasmablast survival. J Immunol. 2006;176:165–172. doi: 10.4049/jimmunol.176.1.165. [DOI] [PubMed] [Google Scholar]
  • 11.Ridings J, Dinan L, Williams R, Roberton D, Zola H. Somatic mutation of immunoglobulin V(H)6 genes in human infants. Clin Exp Immunol. 1998;114:33–39. doi: 10.1046/j.1365-2249.1998.00694.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Lundell AC, Hesselmar B, Nordstrom I, Adlerberth I, Wold AE, Rudin A. Higher B-cell activating factor levels at birth are positively associated with maternal dairy farm exposure and negatively related to allergy development. J Allergy Clin Immunol. 2015;136:1074–1082. e1073. doi: 10.1016/j.jaci.2015.03.022. [DOI] [PubMed] [Google Scholar]
  • *13.Mohanram V, Demberg T, Musich T, Tuero I, Vargas-Inchaustegui DA, Miller-Novak L, Venzon D, Robert-Guroff M. B Cell Responses Associated with Vaccine-Induced Delayed SIVmac251 Acquisition in Female Rhesus Macaques. J Immunol. 2016;197:2316–2324. doi: 10.4049/jimmunol.1600544. Detailed study documenting sex differences in humoral immune responses and protection from SIV in non-human primates. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Tuero I, Mohanram V, Musich T, Miller L, Vargas-Inchaustegui DA, Demberg T, Venzon D, Kalisz I, Kalyanaraman VS, Pal R, et al. Mucosal B Cells Are Associated with Delayed SIV Acquisition in Vaccinated Female but Not Male Rhesus Macaques Following SIVmac251 Rectal Challenge. PLoS Pathog. 2015;11:e1005101. doi: 10.1371/journal.ppat.1005101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Lorenzo ME, Hodgson A, Robinson DP, Kaplan JB, Pekosz A, Klein SL. Antibody responses and cross protection against lethal influenza A viruses differ between the sexes in C57BL/6 mice. Vaccine. 2011;29:9246–9255. doi: 10.1016/j.vaccine.2011.09.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Fan H, Dong G, Zhao G, Liu F, Yao G, Zhu Y, Hou Y. Gender differences of B cell signature in healthy subjects underlie disparities in incidence and course of SLE related to estrogen. J Immunol Res. 2014;2014:814598. doi: 10.1155/2014/814598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *17.Pauklin S, Sernandez IV, Bachmann G, Ramiro AR, Petersen-Mahrt SK. Estrogen directly activates AID transcription and function. J Exp Med. 2009;206:99–111. doi: 10.1084/jem.20080521. Mechanistic understanding of how estrogens affect antibody production in B cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Frasca D, Diaz A, Romero M, Landin AM, Blomberg BB. Age effects on B cells and humoral immunity in humans. Ageing Res Rev. 2011;10:330–335. doi: 10.1016/j.arr.2010.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **19.Gubbels Bupp MR. Sex, the aging immune system, and chronic disease. Cell Immunol. 2015;294:102–110. doi: 10.1016/j.cellimm.2015.02.002. Detailed review about how the immune system ages differently in men than women and the long-term effects on diseases, including autoimmune diseases. [DOI] [PubMed] [Google Scholar]
  • 20.Strindhall J, Skog M, Ernerudh J, Bengner M, Lofgren S, Matussek A, Nilsson BO, Wikby A. The inverted CD4/CD8 ratio and associated parameters in 66-year-old individuals: the Swedish HEXA immune study. Age (Dordr) 2013;35:985–991. doi: 10.1007/s11357-012-9400-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lee OJ, Cho YN, Kee SJ, Kim MJ, Jin HM, Lee SJ, Park KJ, Kim TJ, Lee SS, Kwon YS, et al. Circulating mucosal-associated invariant T cell levels and their cytokine levels in healthy adults. Exp Gerontol. 2014;49:47–54. doi: 10.1016/j.exger.2013.11.003. [DOI] [PubMed] [Google Scholar]
  • 22.Hirokawa K, Utsuyama M, Hayashi Y, Kitagawa M, Makinodan T, Fulop T. Slower immune system aging in women versus men in the Japanese population. Immun Ageing. 2013;10:19. doi: 10.1186/1742-4933-10-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Amadori A, Zamarchi R, De Silvestro G, Forza G, Cavatton G, Danieli GA, Clementi M, Chieco-Bianchi L. Genetic control of the CD4/CD8 T-cell ratio in humans. Nat Med. 1995;1:1279–1283. doi: 10.1038/nm1295-1279. [DOI] [PubMed] [Google Scholar]
  • 24.Hao Y, O'Neill P, Naradikian MS, Scholz JL, Cancro MP. A B-cell subset uniquely responsive to innate stimuli accumulates in aged mice. Blood. 2011;118:1294–1304. doi: 10.1182/blood-2011-01-330530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Rubtsov AV, Rubtsova K, Fischer A, Meehan RT, Gillis JZ, Kappler JW, Marrack P. Toll-like receptor 7 (TLR7)-driven accumulation of a novel CD11c(+) B-cell population is important for the development of autoimmunity. Blood. 2011;118:1305–1315. doi: 10.1182/blood-2011-01-331462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Rubtsov AV, Rubtsova K, Kappler JW, Marrack P. TLR7 drives accumulation of ABCs and autoantibody production in autoimmune-prone mice. Immunol Res. 2013;55:210–216. doi: 10.1007/s12026-012-8365-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *27.Souyris M, Cenac C, Azar P, Daviaud D, Canivet A, Grunenwald S, Pienkowski C, Chaumeil J, Mejía J, Guéry J-C. TLR7 escapes X chromosome inactivation in immune cells. Science Immunology. 2018;3:eaap8855. doi: 10.1126/sciimmunol.aap8855. First documentation that TLR7 has biallelic expression in B cells from women and is associated with development of autoimmune disease. [DOI] [PubMed] [Google Scholar]
  • 28.Fink AL, Klein SL. Sex and Gender Impact Immune Responses to Vaccines Among the Elderly. Physiology (Bethesda) 2015;30:408–416. doi: 10.1152/physiol.00035.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Horstman AM, Dillon EL, Urban RJ, Sheffield-Moore M. The role of androgens and estrogens on healthy aging and longevity. J Gerontol A Biol Sci Med Sci. 2012;67:1140–1152. doi: 10.1093/gerona/gls068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Giefing-Kröll C, Berger P, Lepperdinger G, Grubeck-Loebenstein B. How sex and age affect immune responses, susceptibility to infections, and response to vaccination. Aging cell. 2015;14:309–321. doi: 10.1111/acel.12326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kumru S, Godekmerdan A, Yilmaz B. Immune effects of surgical menopause and estrogen replacement therapy in peri-menopausal women. J Reprod Immunol. 2004;63:31–38. doi: 10.1016/j.jri.2004.02.001. [DOI] [PubMed] [Google Scholar]
  • 32.Kamada M, Irahara M, Maegawa M, Yasui T, Yamano S, Yamada M, Tezuka M, Kasai Y, Deguchi K, Ohmoto Y, et al. B cell subsets in postmenopausal women and the effect of hormone replacement therapy. Maturitas. 2001;37:173–179. doi: 10.1016/s0378-5122(00)00180-8. [DOI] [PubMed] [Google Scholar]
  • 33.Giglio T, Imro MA, Filaci G, Scudeletti M, Puppo F, De Cecco L, Indiveri F, Costantini S. Immune cell circulating subsets are affected by gonadal function. Life Sci. 1994;54:1305–1312. doi: 10.1016/0024-3205(94)00508-7. [DOI] [PubMed] [Google Scholar]
  • 34.Deguchi K, Kamada M, Irahara M, Maegawa M, Yamamoto S, Ohmoto Y, Murata K, Yasui T, Yamano S, Aono T. Postmenopausal changes in production of type 1 and type 2 cytokines and the effects of hormone replacement therapy. Menopause. 2001;8:266–273. doi: 10.1097/00042192-200107000-00008. [DOI] [PubMed] [Google Scholar]
  • 35.Moore SE, Collinson AC, Fulford AJ, Jalil F, Siegrist CA, Goldblatt D, Hanson LA, Prentice AM. Effect of month of vaccine administration on antibody responses in The Gambia and Pakistan. Trop Med Int Health. 2006;11:1529–1541. doi: 10.1111/j.1365-3156.2006.01700.x. [DOI] [PubMed] [Google Scholar]
  • *36.Noho-Konteh F, Adetifa JU, Cox M, Hossin S, Reynolds J, Le MT, Sanyang LC, Drammeh A, Plebanski M, Forster T, et al. Sex-Differential Non-Vaccine-Specific Immunological Effects of Diphtheria-Tetanus-Pertussis and Measles Vaccination. Clin Infect Dis. 2016;63:1213–1226. doi: 10.1093/cid/ciw492. Documentation of sex differences in responses to vaccines in childhood. [DOI] [PubMed] [Google Scholar]
  • 37.McMahon BJ, Williams J, Bulkow L, Snowball M, Wainwright R, Kennedy M, Krause D. Immunogenicity of an inactivated hepatitis A vaccine in Alaska Native children and Native and non-Native adults. J Infect Dis. 1995;171:676–679. doi: 10.1093/infdis/171.3.676. [DOI] [PubMed] [Google Scholar]
  • 38.Michaels RM, Rogers KD. A sex difference in immunologic responsiveness. Pediatrics. 1971;47:120–123. [PubMed] [Google Scholar]
  • 39.Jensen KJ, Ndure J, Plebanski M, Flanagan KL. Heterologous and sex differential effects of administering vitamin A supplementation with vaccines. Trans R Soc Trop Med Hyg. 2015;109:36–45. doi: 10.1093/trstmh/tru184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Martins C, Garly ML, Bale C, Rodrigues A, Benn CS, Whittle H, Aaby P. Measles antibody levels after vaccination with Edmonston-Zagreb and Schwarz measles vaccine at 9 months or at 9 and 18 months of age: a serological study within a randomised trial of different measles vaccines. Vaccine. 2013;31:5766–5771. doi: 10.1016/j.vaccine.2013.08.044. [DOI] [PubMed] [Google Scholar]
  • 41.Jackowska T, Dziadek J, Wasilewski R, Wasik M. [Safety and efficacy of varicella vaccine in healthy children--one year follow-up] Med Wieku Rozwoj. 2008;12:653–659. [PubMed] [Google Scholar]
  • 42.Engler RJ, Nelson MR, Klote MM, VanRaden MJ, Huang C-Y, Cox NJ, Klimov A, Keitel WA, Nichol KL, Carr WW. Half-vs full-dose trivalent inactivated influenza vaccine (2004–2005): age, dose, and sex effects on immune responses. Archives of internal medicine. 2008;168:2405–2414. doi: 10.1001/archinternmed.2008.513. [DOI] [PubMed] [Google Scholar]
  • 43.Vermeiren AP, Hoebe CJ, Dukers-Muijrers NH. High non-responsiveness of males and the elderly to standard hepatitis B vaccination among a large cohort of healthy employees. J Clin Virol. 2013;58:262–264. doi: 10.1016/j.jcv.2013.07.003. [DOI] [PubMed] [Google Scholar]
  • 44.Falsey AR, Treanor JJ, Tornieporth N, Capellan J, Gorse GJ. Randomized, double-blind controlled phase 3 trial comparing the immunogenicity of high-dose and standard-dose influenza vaccine in adults 65 years of age and older. Journal of Infectious Diseases. 2009;200:172–180. doi: 10.1086/599790. [DOI] [PubMed] [Google Scholar]
  • 45.Brandão AP, De Oliveira TC, de Cunto Brandileone MC, Gonçalves JE, Yara TI, Simonsen V. Persistence of antibody response to pneumococcal capsular polysaccharides in vaccinated long term-care residents in Brazil. Vaccine. 2004;23:762–768. doi: 10.1016/j.vaccine.2004.07.024. [DOI] [PubMed] [Google Scholar]
  • 46.Goldblatt D, Southern J, Andrews N, Ashton L, Burbidge P, Woodgate S, Pebody R, Miller E. The immunogenicity of 7-valent pneumococcal conjugate vaccine versus 23-valent polysaccharide vaccine in adults aged 50–80 years. Clinical Infectious Diseases. 2009;49:1318–1325. doi: 10.1086/606046. [DOI] [PubMed] [Google Scholar]
  • 47.Hainz U, Jenewein B, Asch E, Pfeiffer K-P, Berger P, Grubeck-Loebenstein B. Insufficient protection for healthy elderly adults by tetanus and TBE vaccines. Vaccine. 2005;23:3232–3235. doi: 10.1016/j.vaccine.2005.01.085. [DOI] [PubMed] [Google Scholar]
  • 48.Stark K, Schönfeld C, Barg J, Molz B, Vornwald A, Bienzle U. Seroprevalence and determinants of diphtheria, tetanus and poliomyelitis antibodies among adults in Berlin, Germany. Vaccine. 1999;17:844–850. doi: 10.1016/s0264-410x(98)00269-2. [DOI] [PubMed] [Google Scholar]
  • 49.Marlovits S, Stocker R, Efstratiou A, Broughton K, Kaider A, Vecsei V, Kollaritsch H. Effect on diphtheria immunity of combined tetanus and diphtheria booster vaccination in adults. European Journal of Clinical Microbiology and Infectious Diseases. 2000;19:506–513. doi: 10.1007/s100960000305. [DOI] [PubMed] [Google Scholar]
  • 50.Lu FX, Abel K, Ma Z, Rourke T, Lu D, Torten J, McChesney M, Miller CJ. The strength of B cell immunity in female rhesus macaques is controlled by CD8+ T cells under the influence of ovarian steroid hormones. Clin Exp Immunol. 2002;128:10–20. doi: 10.1046/j.1365-2249.2002.01780.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Nguyen DC, Masseoud F, Lu X, Scinicariello F, Sambhara S, Attanasio R. 17beta-Estradiol restores antibody responses to an influenza vaccine in a postmenopausal mouse model. Vaccine. 2011;29:2515–2518. doi: 10.1016/j.vaccine.2011.01.080. [DOI] [PubMed] [Google Scholar]
  • 52.Pap PL, Czirjak GA, Vagasi CI, Barta Z, Hasselquist D. Sexual dimorphism in immune function changes during the annual cycle in house sparrows. Naturwissenschaften. 2010;97:891–901. doi: 10.1007/s00114-010-0706-7. [DOI] [PubMed] [Google Scholar]
  • 53.Fargallo JA, Martinez-Padilla J, Toledano-Diaz A, Santiago-Moreno J, Davila JA. Sex and testosterone effects on growth, immunity and melanin coloration of nestling Eurasian kestrels. J Anim Ecol. 2007;76:201–209. doi: 10.1111/j.1365-2656.2006.01193.x. [DOI] [PubMed] [Google Scholar]
  • 54.Zuk M. The sicker sex. PLoS Pathog. 2009;5:e1000267. doi: 10.1371/journal.ppat.1000267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **55.Vermillion MS, Klein SL. Pregnancy and infection: using disease pathogenesis to inform vaccine strategy. NPJ Vaccines. 2018;3 doi: 10.1038/s41541-017-0042-4. A comprehensive review of infection and protection of mother and offspring during pregnancy, with consideration of antibody transfer from mother to young. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES