Skip to main content
Journal of Virology logoLink to Journal of Virology
. 2019 Oct 15;93(21):e00797-19. doi: 10.1128/JVI.00797-19

Host Factors Impact Vaccine Efficacy: Implications for Seasonal and Universal Influenza Vaccine Programs

Santosh Dhakal a, Sabra L Klein a,b,
Editor: Carolyn B Coynec
PMCID: PMC6803252  PMID: 31391269

Influenza is a global public health problem. Current seasonal influenza vaccines have highly variable efficacy, and thus attempts to develop broadly protective universal influenza vaccines with durable protection are under way. While much attention is given to the virus-related factors contributing to inconsistent vaccine responses, host-associated factors are often neglected.

KEYWORDS: aging, microbiota, obesity, pandemic, pregnancy, seasonal influenza, sex difference

ABSTRACT

Influenza is a global public health problem. Current seasonal influenza vaccines have highly variable efficacy, and thus attempts to develop broadly protective universal influenza vaccines with durable protection are under way. While much attention is given to the virus-related factors contributing to inconsistent vaccine responses, host-associated factors are often neglected. Growing evidences suggest that host factors including age, biological sex, pregnancy, and immune history play important roles as modifiers of influenza virus vaccine efficacy. We hypothesize that host genetics, the hormonal milieu, and gut microbiota contribute to host-related differences in influenza virus vaccine efficacy. This review highlights the current insights and future perspectives into host-specific factors that impact influenza vaccine-induced immunity and protection. Consideration of the host factors that affect influenza vaccine-induced immunity might improve influenza vaccines by providing empirical evidence for optimizing or even personalizing vaccine type, dose, and use of adjuvants for current seasonal and future universal influenza vaccines.

INTRODUCTION

Each year, seasonal influenza epidemics cause 3 to 5 million severe cases of respiratory illnesses and 290,000 to 650,000 deaths worldwide (1). Such epidemics generally occur during the winter months when lower temperature and humidity favor virus dissemination. Although all people can acquire influenza viruses, children, the elderly, pregnant women, and individuals with either chronic medical conditions or immunosuppression are at higher risk (2). The influenza A virus (IAV) H1N1 subtype dominated in the early 20th century, the H2N2 subtype of IAV was predominant in the late 1950s and 1960s, and H1N1 and H3N2 subtypes of IAV and different lineages of influenza B viruses have been responsible for seasonal epidemics in last several decades (2, 3). Influenza A and B viruses frequently acquire point mutations on their surface glycoproteins (antigenic drift), resulting in variable levels of morbidity and mortality each season (2). In the United States, seasonal influenza epidemics since 2010 and 2011 have resulted in between 9.3 and 49 million cases, between 140,000 and 960,000 hospitalizations, and between 12,000 and 79,000 deaths annually (4). In the 2017–2018 season, influenza-related hospitalizations in the United States reached almost a million, with nearly 79,000 deaths, of which older adults (≥65 year of age) were affected most severely (4). In addition to annual epidemics, influenza pandemics occur sporadically at unpredictable frequencies, with four such pandemics occurring in last century (i.e., 1918, 1957, 1968, and 2009), which resulted in millions of deaths worldwide (2). Pandemic influenza viruses are generated by reassortment of genetic segments (antigenic shift) between two or more influenza viruses in swine or avian hosts. The resulting virus is antigenically different than seasonal influenza strains and causes severe illness and high mortality in the population, which lacks sufficient preexisting immunity (2). Sporadic human infections have also been recorded in last few decades with novel avian origin IAVs (e.g., H5N1 and H7N9) and swine origin variant viruses (H3N2), which may undergo sufficient mutations and result in pandemics if sustained human to human transmission occurs (5, 6).

Vaccination is the most effective means to prevent influenza infection. Influenza vaccines have been in use since the 1940s in the United States (2). Most commonly used influenza vaccines are inactivated influenza virus (IIV) vaccines administered through the intramuscular route with the exception of intranasal live-attenuated influenza vaccines (LAIVs) recommended for use in young children (2). Current seasonal influenza vaccines contain two IAVs (H1N1 and H3N2) and one (in the trivalent influenza vaccine [TIV]) or two (in quadrivalent influenza vaccine [QIV]) influenza B viruses to increase the breadth of immunity (7). The immunity conferred by these vaccines is specific to the vaccine antigen and wanes over time (8); thus, annual vaccination is recommended. Currently, the strains to be used in the annual influenza vaccines are decided based on the surveillance report of World Health Organization (WHO) laboratories established throughout the world (7). To prepare seasonal influenza vaccines, the hemagglutinin (HA) and neuraminidase (NA) gene segments of the selected strains are reassorted into the replication efficient A/Puerto Rico/08/1934 (H1N1) IAV, and the virus is grown in embryonated chicken eggs, followed by chemical inactivation (2). LAIV, which is delivered as a nasal spray, was launched in the United States in 2003 targeting the pediatric population (9). In 2016, the use of LAIV was suspended in the United States owing to poor effectiveness particularly against A(H1N1)pdm09 virus (9); other countries, however, including the United Kingdom and Finland continued its use and even the Advisory Committee on Immunization Practices (ACIP) in the United States recommended reintroduction of LAIV vaccine for the 2018/19 influenza season (9, 10).

Influenza vaccination is recommended in the United States for any individuals over 6 months of age who do not have any contraindications (9). Pregnant women, health care workers, children, the elderly, and immunocompromised individuals are prioritized for vaccination because the risk of severe disease is higher among them. Seasonal influenza vaccines, in general, reduce the risk of influenza infection and severe outcomes in individuals of any age compared with unvaccinated individuals (11). The US Centers for Disease Control and Prevention (CDC) estimated that vaccination averted around 5.29 million influenza-related illnesses, 2.64 million medical visits, and 84,700 hospitalizations in the United States during 2016–2017 influenza season (11). The protective benefit of seasonal influenza vaccines is mainly based on the ability to produce antibodies against HA, the major surface glycoprotein, of the virus. Such hemagglutination inhibition (HAI) antibodies bind to the head region of HA antigen and prevent the virus from binding to the receptor on respiratory epithelial cells (12). The HAI titer is the most commonly used correlate of protection for seasonal influenza vaccines where a titer of ≥40 is considered protective (13, 14). The seasonal influenza vaccines, however, provide only strain-specific protection and limited or no protection against novel strains with pandemic potential; the requirement of annual reformulation of vaccines results in vaccine strain selection being months behind in the arm race with the constantly evolving influenza viruses (5). Egg adaptation can result in additional mutations in the influenza vaccine strains which impair the functional ability of antibodies produced against the vaccine virus to neutralize circulating viruses further reducing influenza vaccine efficacy (15). In general, the effectiveness of seasonal influenza vaccines is highly variable from season to season and ranges between 10 and 60% (5).

As a result of variability in seasonal influenza vaccine efficacy, there is growing interest in the development of universal influenza vaccines that can provide broader and longer duration immunity. As per the strategic plan of National Institute of Allergy and Infectious Diseases (NIAID) to improve current influenza vaccines, the universal influenza vaccines should have at least 75% effectiveness, protect against both group 1 and 2 of IAVs, confer multiseason protection, and be effective in all age groups (5). Different strategies are being considered to develop universal influenza vaccines, including chimeric or headless HA-based approaches that induce broadly protective HA stalk-specific antibodies (16), matrix protein 2 (M2) targeting vaccines that elicit antibodies against the highly conserved M2 protein with cross-protective properties (17), and T cell-stimulating vaccines based on conserved internal proteins, such as the nucleoprotein (NP) and matrix protein 1 (M1) (18). Although considerable attention is being paid to the rational design of improved influenza vaccines, including universal influenza vaccines, continued attention should be paid to the host-related factors, including age, sex, pregnancy, immune history, and obesity (Fig. 1), that can alter vaccine efficacy. Furthermore, little consideration has been given to the targetable mechanisms, including the composition of the gut microbiota, genetics, and even hormones that can contribute to how age, sex, pregnancy, and even obesity can impact influenza vaccine-induced immunity and protection.

FIG 1.

FIG 1

Several host-associated factors can impact influenza vaccine efficacy (i.e., the ability of vaccine-induced immunity to protect against subsequent influenza virus infection). Studies of humans and nonhuman animals immunized with inactivated influenza vaccines (IIVs) suggest that biological sex and gender-associated factors (e.g., behavior), age, and immune history are predictors of IIV-induced humoral immunity (i.e., correlate of protection) and presumably protection against influenza. Evidence also suggests that the composition of the gut microbiota, obesity, and pregnancy can impact IIV-induced immunity in humans. With growing concerns about the efficacy of seasonal IIVs, there is a push for development and implementation of universal influenza vaccine platforms. The impact of host factors, in both preclinical and clinical studies, on vaccine-induced immunity should be considered in an effort to maximize immunity conferred by influenza vaccines, including universal influenza vaccines.

EFFECTS OF AGE ON INFLUENZA VACCINE OUTCOME

Influenza vaccination in children above 6 months of age has been recommended since the 2002–2003 influenza season in the United States. Immunogenicity studies show that full-dose vaccination (i.e., two-dose vaccine at 4-week interval, if not vaccinated before) induces a protective level of serum antibodies (HAI  ≥ 40) in children (19). One study using TIV in vaccine-naive and seronegative 5- to 8-year-old children showed that 85, 68, and 48% of the children develop protective antibody response (HAI ≥  40) against H1N1, H3N2, and influenza B virus strains, respectively, after receiving two doses of vaccine (20). In the case of LAIV, mucosal antibody and cell-mediated immune responses are associated with protective efficacy in children, but specific correlates of protection for LAIV vaccines are not well established (21). Randomized controlled trials have shown that the efficacy of IIVs in children ranges from 43 to 91%, whereas for LAIV, the efficacy is 64 to 93% (22).

A meta-analysis of vaccine efficacy studies in 10 different randomized trials In adults (18 to 65 years) showed an average pooled efficacy of 59% for TIV (23), with QIV resulting in seroprotection (HAI ≥ 40) and seroconversion (i.e., 4-fold increase in HAI titer postvaccination) rates that are comparable or greater than TIV in young adults (24). In general, the clinical vaccine efficacy estimates in elderly (≥65 years of age) are lower than in young adults (<65 years of age) (25). A review of vaccine antibody response studies from 1986 to 2002 showed that the seroconversion rates were higher in young adults compared to aged adults against H1N1 (60% versus 42%), H3N2 (62% versus 51%), and influenza B (58 versus 35%) viruses (25). Likewise, seroprotection rates were also greater in young adults than in aged adults against H1N1 (83% versus 69%), H3N2 (84% versus 74%), and influenza B (78% versus 67%) viruses (25). Vaccine effectiveness (based on observational studies) is similar among children, young adults, and aged adults against A/H1N1 and influenza B viruses; whereas vaccine effectiveness is highest in children, lower in young adults, and lowest in aged adults against A/H3N2 (26). Influenza vaccine effectiveness in the UK during 2016/17 season also showed a declining trend of vaccine effectiveness in aged adults against all influenza virus antigens compared to younger adults (27). In general, influenza vaccine-induced immunity declines with age, which is likely affected by host factors, such as age-associated immunosenescence and immune history, as well as virus factors, including antigenic drift.

Regarding vaccine safety, influenza vaccines in general are safe and well tolerated by different age groups with rare serious or clinically important adverse events (28). Mild and transient local and systemic reactions to influenza vaccines are reported in all age groups, with comparatively higher adverse reactions being observed in younger subjects (29, 30). The adverse reactions to influenza vaccines may vary depending upon the nature of the vaccine (e.g., IIV or LAIV), vaccine composition (selection of antigens and adjuvants), route of administration (intramuscular, intranasal, or others), and the dose of vaccines. Hence, close monitoring of the safety profile is necessary across different age groups while testing novel vaccine platforms in clinical studies.

The reduced efficacy of influenza vaccine in aged humans has been associated with age-dependent changes in innate and adaptive immune function due to immunosenescence. Lower expression of CD80 costimulatory molecules in activated monocytes (31), a reduced number of plasmacytoid dendritic cells and a generalized decrease in Toll-like receptor 7 (TLR7)- and TLR8-induced cytokines (alpha interferon [IFN-α] and interleukin-12p40 [IL-12p40]) in dendritic cells (32), and impaired inflammatory responses in aged adults due to elevated levels of the anti-inflammatory cytokine IL-10 (33) are associated with lower seroconversion and seroprotection rates of vaccines in older adults compared to young adults. Systems biology approaches have found the association of higher baseline innate inflammatory responses in aged adults with diminished antibody responses postimmunization with TIV over five consecutive influenza seasons (34). Reduced influenza-specific activation-induced cytidine deaminase (AID) responses and the percentage of switch memory B cells (35, 36), reduced affinity maturation (37), lower number of antibody-secreting cells or plasmablasts (38), impaired memory B cell to plasma cell differentiation (39), diminished antibody diversity (40), and reduced plasticity of B cell receptor repertoires (41) are likely to be the B-cell-associated defects associated with inefficient antibody responses after influenza vaccination in aged adults. Shortening of telomere length in B cells is also linked to lower antibody production and inefficient CD8+ T cell proliferation in aged adults (42). The loss of CD28 molecules that are pivotal in T cell activation and germinal center development and accumulation of CD8+ CD28 T cells (43, 44), a decline in influenza-specific memory T cells (45), a reduced frequency and function of circulatory T follicular helper cells (46), and age-dependent decline in granzyme-, perforin-, and IFN-γ-mediated cytolytic activity of CD8+ T cells (47, 48) represent some of the T cell events in aged adults that may contribute to lower vaccine-induced immunity and protection. The expression of the proliferative senescence marker, KLRG1, and the inhibitory receptor CD57 on CD8+ T cells prevaccination can also predict lower antibody production in aged adults (49). Infection with cytomegalovirus, which is more prevalent in older subjects, also affects influenza vaccine-induced immunity in aged individuals (50). A recent study has shown that plasmablasts from older individuals have reduced somatic hypermutation of immunoglobulin variable genes, resulting in limited adaptability of antibody responses to drifted epitopes of influenza virus and highlighting the fact that a better vaccine for older individuals should focus on enhancing the antibody affinity maturation process (51). Studies in aged humans also show that high-dose vaccine and adjuvant supplementation improves the performance of inactivated vaccines compared to the standard-dose vaccination, with these strategies now incorporated in seasonal influenza vaccine campaigns for older individuals (52, 53).

Studies in mice also reveal that immunization of aged mice (>16 months) with either monovalent or trivalent IIVs induces lower antibody responses and protection compared to young mice (2 to 3 months) (5456). Such differences are in part associated with age-dependent changes in the hormonal milieu, cytokine profile, and antigen-presenting cell responsiveness after immunization (5456). Influenza nucleoprotein-based vaccine studies also indicate that aged mice have alterations in CD4+ T-helper and CD8+ cytotoxic T cell frequencies compared to young mice (57, 58). Aged mice also require multiple doses of vaccines, higher quantities of antigen, or the addition of adjuvant to elicit improved immune responses and protective efficacy of influenza vaccines compared to young mice (59, 60).

In mice, IAV NP and M2-based universal influenza vaccine candidates tested across different age groups indicate that antigen-specific antibody and T cell responses decline with age, leading to a reduced protective efficacy in older mice (61). Studies in humans and mice highlight the necessity of including age in preclinical and clinical studies of seasonal and universal influenza vaccines.

SEX DIFFERENCES IN INFLUENZA VACCINE EFFICACY

Biological sex (i.e., being male or female based on sex chromosome complement, gonadal tissues, and sex steroid concentrations) and gender (i.e., sociocultural influences that affect roles, behaviors, and activities that are deemed appropriate for men or women) are associated with the influenza vaccine acceptance rate, the reporting of adverse reactions, and the development of immune responses postvaccination (62). The influenza vaccine acceptance rate is reportedly lower, whereas passive reporting of local and systemic adverse reaction is higher, in females than males (62). In young adults (18 to 49 years), immunization with seasonal TIV vaccine resulted in at least two times greater HAI antibodies in females than males (63). Administration of a half-dose TIV in females induced antibody responses to the H1N1, H3N2, and influenza B antigens that are almost equivalent to the full-dose vaccine response in males (63). Higher neutralizing antibodies are observed regardless of age in females after either monovalent vaccination or TIV, with the differences being reported against both influenza A and B viruses (56, 63, 64). Among aged individuals (≥65 years), both the seroconversion and the seroprotection rates are higher in females compared to males after immunization with TIV (65). A greater HAI antibody response in aged adult females is also observed against H1N1 and H3N2 viruses after immunization with high-dose seasonal TIV (66). Vaccination with a monovalent, unadjuvanted H1N1 pandemic 2009 vaccine resulted in females having significantly higher seroprotection and seroconversion than males in Taiwan (67). Likewise, aged females have significantly higher postvaccination HAI titers than similarly aged males after a single dose of unadjuvanted 2009 H1N1 pandemic vaccination in some (68) but not all (56) studies in the United States. The impact of biological sex on immune responses or adverse reactions after the receipt of either QIV or LAIV has not been reported in humans.

Limited human studies have considered the effects of biological sex on vaccine effectiveness studies but have consistently shown that influenza-vaccinated females have a lower risk of hospitalizations and deaths compared to vaccinated males (6971). A recent study evaluated vaccine effectiveness across seven seasons in Canada and showed that overall vaccine effectiveness was higher in females than in males; the difference was more pronounced in response to A/H3N2 and influenza B virus than A/H1N1 (72), with the observed sex difference being more pronounced in older (≥50 years of age) than in younger (<20 years of age) individuals (72). These findings highlight the need to include sex as a potential modifier of influenza vaccine outcome in future vaccine effectiveness studies.

Studies in mice further illustrate sex differences in immunity and protection with influenza vaccines. After primary inoculation with a sublethal dose of H1N1 or H3N2 influenza virus, adult female C57BL/6 mice generated higher neutralizing and total anti-influenza virus antibodies (73). After secondary challenge, males and females show similar levels of protection against homologous virus, but females have better protection against heterosubtypic viruses, indicating that females developed better cross-protective immunity than males (73). After vaccination with either whole inactivated IAV, TIV, or QIV, adult female mice generate greater quantity and quality of influenza-specific antibody responses than do males (7476). Antibody derived from vaccinated females also is better at protecting both naive males and females than antibody from that from males, and this protection is associated with increased antibody specificity and avidity to the H1N1 virus (76). The TLR7 gene (Tlr7) is encoded on the X chromosome, is also expressed in B cells, and plays a role in isotype switching (77). The expression of Tlr7 is greater in B cells from vaccinated females than in B cells from males and is associated with reduced DNA methylation in the Tlr7 promoter region, a higher neutralizing antibody, class switch recombination, and antibody avidity in females (76). Deletion of Tlr7 reduced sex differences in vaccine-induced antibody responses and protection after challenge and had a greater impact on responses in females than males. Taken together, these data illustrate that greater TLR7 activation in B cells and antibody production in females improves the efficacy of IIVs against influenza.

Global gene expression analysis of B cells from healthy human adults also indicates a differential expression of genes between male and females, particularly those that contain estrogen response elements in their promoter regions, indicating that hormone signaling may regulate gene expression in B cells (78). In mice, 17β-estradiol is positively associated with IAV neutralizing antibody production in females, indicating the role of estrogen in modulating influenza vaccine-induced immunity in females of reproductive age (56, 79). In humans, the lower neutralizing antibody response in males compared to females after TIV vaccine administration is associated with a higher level of serum testosterone and greater lipid metabolism (64).

To date, no animal studies of universal influenza vaccines have examined sex differences in vaccine-induced immunity or protection. To gain insight into the consideration of biological sex in universal influenza vaccine studies in animal models, we performed a literature search in PubMed using the keywords “universal influenza vaccine” for the year 2018. This search resulted in 42 influenza vaccine studies in different animal models, with 86% (36/42) of them using only female animals; 7% (3/42) using both sexes, but not disaggregating results based on sex; and the remainder (7% [3/42]) either using only male animals or not reporting the sex of the animals. To date, preclinical studies have failed to acknowledge the importance of biological sex in vaccine-induced immunity and protection.

EFFECTS OF IMMUNE HISTORY ON INFLUENZA VACCINE EFFICACY

Immune history is acquired over time through both virus exposures and vaccination, which affects the quality and quantity of antibody developed against influenza viruses later in life. Early life exposure to influenza viruses that occurs within the first decade of life presumably dominates the development of influenza-specific antibody responses later in life (80, 81). This phenomenon is known as “original antigenic sin” (OAS) and was put forward by Thomas Francis, Jr., in the 1960s (82). Currently, the concept of OAS is also referred as “immune imprinting” to address both the positive and the negative aspects of immune history on influenza virus vaccine efficacy (80). Immune imprinting facilitates the activation of memory B cells over de novo activation of naive B cells, thereby establishing a hierarchy of antibody responses where the highest response is generated against the strains from childhood, with subsequent strains inducing lower titers of antibody (80).

A cross-sectional study in China showed that neutralizing antibodies remained highest against the H3N2 viruses that circulated in the first decade of participants life, with lower neutralizing antibody responses observed against other H3N2 strains that circulated in subsequent years (83). Similarly, a longitudinal study over a 20-year period indicated that neutralizing antibodies against previously encountered influenza virus strains expand continuously over time (84). High-throughput studies of human plasmablasts induced by vaccination suggest that influenza vaccination induces preferential recall of memory B cells specific to influenza virus strains that circulated in previous years compared to the strains used for vaccination in more recent years (85, 86). Immune imprinting can also be replicated in the laboratory using sequential influenza virus infections of mice, rats, or ferrets (8789). A study in mice, for example, showed that the effect of immune imprinting is more pronounced if the first exposure to IAV is through infection rather than vaccination (89).

The differences in the quality or cross-reactivity of antibody responses after influenza vaccination in different age cohorts is also partly explained by the differences in imprinting to viruses associated with birth year (90). Higher influenza virus susceptibility in older individuals may be caused by early life immune imprinting altering antibody responses against drifted influenza viruses later in life, despite the high immunization rate within this population (91). As a result of a mutation in the HA of the circulating H1N1 IAV during the 2013–2014 influenza season, the infection rate was unusually high in the middle-aged population, partly because these individuals developed higher antibody titers against the K166 HA of the H1N1 viruses that circulated during their birth years (1965 to 1979) than against the drift variant (K166Q) that was currently in circulation (92). A similar reduction in vaccine effectiveness in older and middle-aged people in the United States and Canada was observed during the H1N1-dominated 2015–2016 influenza season, which also could be associated with immune imprinting to sufficiently different H1N1 strains (93, 94). Mouse studies have shown that the adverse effect of immune imprinting can be avoided by including adjuvants in vaccine formulation or through repeated immunizations (95). Human studies suggest that an adjuvanted seasonal influenza vaccine is more effective at inducing antibody responses than a comparable unadjuvanted seasonal influenza vaccine, at least in an aged human population (53, 96).

Immune imprinting can provide a protective benefit during influenza virus infection later in life. One such example is the 2009 influenza pandemic, in which aged individuals with prior immune history with the pandemic 1918 H1N1 influenza virus were less susceptible to 2009 H1N1 than were younger age individuals (97). Likewise, the lower susceptibility observed in older than in younger individuals (i.e., people 18 to 49 years of age) during the 1918 influenza pandemic is hypothesized to be due to exposure to earlier strains of H1 viruses in childhood (98). Using statistical modeling, childhood imprinting with H1N1 and H3N2 virus can also provide protection against H5N1 and H7N9 influenza viruses, respectively, later in life (99). Immune imprinting boosts memory B cell responses that produces broadly neutralizing HA stalk-specific antibodies (80).

Despite reports of both positive and negative effects of immune imprinting on influenza vaccine efficacy, the extent to which immune imprinting is induced by different types of exposures, including different vaccine platforms, is not well characterized. Few studies have examined the role of host factors, including the combined effect of sex and age, in influencing the strength and magnitude of immune imprinting both in humans and nonhuman animals.

PREGNANCY AND INFLUENZA VACCINE EFFICACY

Pregnancy is associated with physiological and immunological alterations intended to maintain an optimal environment for the growing fetus. Hormones, including estradiol and progesterone, which are immunomodulatory, vary considerably during different stages of pregnancy (100). Cytokines that mediate inflammatory or anti-inflammatory responses and immune cells associated with innate and adaptive immune systems also fluctuate during pregnancy (100, 101). Together, these pregnancy-associated physiological and immunological changes contribute to immunological shifts in pregnant compared to nonpregnant females.

In the case of influenza, pregnant females are at higher risk of influenza virus infection compared to nonpregnant females (102). Considering the greater risk of influenza infection and outcomes, the WHO has prioritized influenza vaccination for pregnant women (103). Immunization during pregnancy serves the dual function of protecting the health of mothers and infants. Because no influenza vaccines are recommended before 6 months of age, neonatal protection from influenza relies on passively transferred antibodies from the mother (103). A study in the United States using TIV showed comparable seroconversion rates when females were vaccinated during either the first or the third trimester of pregnancy (104). As in other populations, a history of prior immunization and greater baseline antibody titers are associated with lower seroconversion rates in pregnant women (104). IIVs result in comparable seroprotection and seroconversion rates between pregnant and nonpregnant women (105, 106). The randomized controlled trials have shown highly variable efficacy of TIV during pregnancy. One study in Nepal showed only 19% efficacy of TIV in preventing influenza-like illnesses in pregnant women, while another study in South Africa observed a 50.4% vaccine efficacy (107, 108). The efficacy of maternal influenza immunization in preventing laboratory-confirmed influenza infection in infants also varies from 30 to 63% in different clinical trials (107109).

Multiple studies show that maternal immunization with IIVs is not associated with increased risk of adverse events in maternal or fetal health (110, 111). A study in 2017 showed that women who received the inactivated pandemic H1N1 2009 vaccine during the first trimester of pregnancy had a greater risk of miscarriage if they had received the same vaccine in the previous year (112). Though this finding does not indicate a causal relationship, it highlights the importance of continued active surveillance in pregnant women and infants, which are all too often not included in clinical trials of drugs or biologics, including vaccines (113).

Studies in mouse models of pregnancy have shown that disruption of cytokine and hormonal pathways, as well as alterations of placental and respiratory pathophysiology, is responsible for the adverse effects of influenza infection during pregnancy, which are mediated by infection-induced suppression of circulating progesterone (114, 115). Influenza vaccination during pregnancy in mice suggests greater protection of neonates through maternal immunization (116, 117). To date, no influenza vaccine has been designed to specifically target pregnant females. In order to develop safe, immunogenic, and highly efficacious vaccines including universal influenza vaccines, pregnancy-associated changes and their impact on vaccine-induced immunity should be considered in experimental studies.

OBESITY AS A COMORBIDITY IN INFLUENZA VACCINE EFFICACY

Obesity is an independent risk factor for influenza-related illnesses and hospitalizations (118). Obesity also impacts the effectiveness of influenza vaccines. Obese adults immunized with TIV, despite developing efficient antibody responses, are at two times greater risk of developing influenza or influenza-like illnesses (119). The relatively lower effectiveness of influenza vaccines in obese individuals is hypothesized to be mediated by insufficient T cell function, since peripheral blood mononuclear cells from TIV-vaccinated obese adults have decreased activation of cytotoxic T cells and reduced expression of functional markers, including IFN-γ and granzyme B (120). Influenza vaccination is still important in obese populations since vaccinated obese children are three times less likely to acquire PCR-confirmed influenza and miss significantly fewer school days compared to their unvaccinated obese counterparts (121). At least one study using diet-induced obesity in adult male mice illustrates that the induction of chronic inflammation might be responsible for reduced efficacy of inactivated monovalent influenza vaccine in obese mice (122). The impact of obesity on immune responses and protection after influenza virus challenge could not be reversed by increasing the dose of vaccine antigen or using an adjuvant in obese mice (123); therefore, further studies are needed to explore the underlying mechanisms for reduced protection against influenza virus infection in obese individuals which will guide the development of an effective influenza vaccine. The microbiome of lean and obese individuals differs considerably in terms of richness and diversity (124) and, given the role of the microbiome in host immunity (125), studies should dissect the interaction of the microbiome with obesity on influenza vaccine effectiveness.

HYPOTHESIZED HOST-RELATED MECHANISMS AFFECTING INFLUENZA VACCINE EFFICACY

Microbiota.

In murine models of influenza infection, the commensal microbiota plays important role in the establishment of proper innate and adaptive immune responses. After antibiotic treatment or in germfree mice, the innate and adaptive immunity to influenza virus is severely compromised compared to specific pathogen-free mice, but the sexes and ages of these mice are inconsistently reported in these studies (126, 127). The intestinal microbiota can regulate the TLR7 pathway after influenza virus infection, at least in adult female mice (128). Several studies in mice have reported the beneficial prophylactic effect of oral and intranasal administration of probiotics, such as Lactobacillus, on influenza virus-specific innate and adaptive immune responses (129, 130). The immunological benefits of adding probiotics into HA- and M2e-based influenza vaccine formulations have also been shown in mice (131, 132). A randomized controlled study in adult humans in Italy showed that those who consumed probiotics before receipt of the seasonal TIV had significantly higher vaccine-specific antibody responses compared to placebo controls (133). Similarly, in the United States, healthy adults that consumed probiotics for 28 days postvaccination with TIV had significantly improved seroprotection against the H3N2 virus strain compared to placebo controls (134). The beneficial effects of probiotics on antibody responses to TIV are also observed in older adults who consumed probiotics continuously either before or after immunization (135, 136).

TLR5, which senses bacterial flagellin produced by the microbiota, is associated with improved efficacy of influenza vaccines. In healthy adults immunized with TIV, there is an association between early expression of TLR5 in blood with the subsequent induction of vaccine-specific antibody responses (137). Because TIV alone does not stimulate TLR5, such activation is likely mediated by other factors, including the host commensal microbiota. A follow- up study in mice that did not indicate either the sexes or the ages of the mice revealed that TLR5-mediated sensing of flagellin produced by intestinal microbiota is necessary for efficient antibody production after influenza vaccination (138). Antibody responses to an IIV also were severely impacted in germfree or antibiotic-treated wild-type mice, as well as in TLR5 knockout mice after immunization with TIV. The immunity was restored in germfree and antibiotic-treated mice after oral administration of a flagellated strain of Escherichia coli or after coinjection of flagellin and TIV (138). This study highlights the influence of microbiota on influenza vaccine responses. Additional studies are necessary to compare how intestinal and respiratory microbiota are associated with influenza virus infection and immunity. Furthermore, most studies evaluating the impact of the microbiota on influenza vaccine-induced immunity do not consider the sex, age, body mass, or reproductive status of the subject and may miss important associations that could explain some variability in the impact of the microbiota on vaccine efficacy.

Genetics.

Host genetic factors play an important role in influenza pathogenesis and immunity to vaccines. Single nucleotide polymorphisms of genes associated with human leukocyte antigen (HLA) molecules, cytokines, and cytokine receptors influence the humoral immune responses to influenza virus vaccines (139). For example, polymorphisms in the IFN-inducible transmembrane protein 3 (IFITM3) gene are associated with increased risk of influenza virus infection, and recent meta-analyses indicate that the polymorphism in IFITM3 is associated with an increased risk of severe influenza in Asians and Caucasians but not in other racial or ethnic groups (140, 141). The IFITM gene, which is induced by IFNs, mediates antiviral response and can also shape adaptive immunity by protecting the survival of resident memory cytotoxic T cell population (142). In adult humans, the association of host genetics and antibody responses to seasonal TIV revealed 20 genes that mediate antibody responses to TIV, including genes related to antigen processing and intracellular trafficking (143). Another study suggested an association between the presence of certain HLA class II alleles in older aged individuals with a higher seroprotective response after immunization with TIV (144). A study by Avnir et al. showed that a phenylalanine (F)-to-leucine (L) polymorphism in the immunoglobulin heavy-chain variable locus (IGHV1-69) can modulate B cell clonal expansion, somatic hypermutation, and neutralizing antibody responses after H5N1 influenza vaccination (145). Vaccinees bearing F alleles develop higher stem-directed broadly neutralizing antibody and microneutralization antibody responses versus individuals carrying L alleles (145). A better understanding of the relationship of genetic variations and influenza vaccine-induced immunity is necessary to predict effectiveness of seasonal or universal influenza vaccine responses, as well as how genetics and even epigenetics may explain the impact of host demographic variables on vaccine-induced immunity.

Hormones.

Sex steroids, mainly estrogens and progesterone in females and testosterone in males, fluctuate over the life course and mediate a wide range of immune responses during infection or vaccination by interacting with their respective receptors expressed in cells of the innate and adaptive immune system (146). Estradiol can directly upregulate AID and induce somatic hypermutation and class switch recombination of immunoglobulins, which indicate its potential role in antibody production and function (147). In adult female mice, ovariectomized mice have significantly lower influenza-specific antibody responses than gonad-intact females after immunization with monovalent inactivated vaccine, which is restored after administration of estrogen to ovariectomized females (56, 79). The rapid decline of estradiol in women after menopause, associated with health disorders including osteoporosis and atherosclerosis, can be managed by hormonal replacement therapy (HRT). HRT that consisted of equine estradiol and medroxyprogesterone reduces baseline the concentration of proinflammatory cytokine and increases the numbers of circulating B cells in postmenopausal women (148, 149). Plasma estradiol levels in postmenopausal women, including those on HRT, are positively correlated with the fold increase in influenza-specific IgG antibody titers after immunization with TIV (150). In both humans and mice vaccinated with a monovalent 2009 H1N1 inactivated vaccine, serum estradiol levels are positively correlated with neutralizing antibody titers in both young and aged adult females (56).

Circulating testosterone concentrations decline gradually in men as they age, and one previous study has indicated that men with higher serum testosterone concentrations have lower antibody responses to TIV (64). In both humans and mice vaccinated with a monovalent 2009 H1N1 inactivated vaccine, higher serum testosterone concentrations are negatively associated with neutralizing antibody titers in young adult but not aged adult individuals (56). In mice, removal of the gonads, and hence the production of estrogens and progesterone in females and testosterone in males, eliminates sex differences in IIV-induced immunity, which can be reversed by the replacement of estradiol in females and testosterone in males (56). Further studies into how sex steroids mediate the effects of sex, age, and pregnancy are required, especially in studies of universal influenza vaccine platforms.

CONCLUSIONS

The data pertaining to seasonal IIVs suggest that the “one size fits all” approach of vaccine administration does not necessarily protect equally across distinct ages, sexes, reproductive periods, or comorbid conditions, with commensal microbial, genetic, and hormonal mechanisms contributing to this variability. As we move closer toward the goal of development of universal influenza vaccines that provides efficient protection across all age groups, host factors must be given greater consideration. Preclinical, clinical, and epidemiological studies must continue to disaggregate and explore the influence of host-specific factors on influenza vaccine-induced immunity and protection. Through greater consideration of the host factors reviewed, either alone or in combination, we may mitigate disparities in influenza vaccine efficacy and develop safer and more efficacious universal influenza vaccines.

ACKNOWLEDGMENTS

The writing of this review was supported by the NIH/NIAID Center of Excellence in Influenza Research and Surveillance contract HHS N272201400007C and the NIH/ORWH/NIA Specialized Center of Research Excellence in Sex Differences U54AG062333.

REFERENCES

  • 1.WHO. 2018. Influenza (seasonal) fact sheet. World Health Organization, Geneva, Switzerland: https://www.who.int/news-room/fact-sheets/detail/influenza-(seasonal). Accessed 5 May 2019. [Google Scholar]
  • 2.Krammer F, Smith GJD, Fouchier RAM, Peiris M, Kedzierska K, Doherty PC, Palese P, Shaw ML, Treanor J, Webster RG, Garcia-Sastre A. 2018. Influenza. Nat Rev Dis Primers 4:3. doi: 10.1038/s41572-018-0002-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Zimmer SM, Burke DS. 2009. Historical perspective: emergence of influenza A (H1N1) viruses. N Engl J Med 361:279–285. doi: 10.1056/NEJMra0904322. [DOI] [PubMed] [Google Scholar]
  • 4.CDC. 2018. Disease burden of influenza fact sheet. Centers for Disease Control and Prevention, Atlanta. GA: https://www.cdc.gov/flu/about/burden/index.html. Accessed 31 December 2018. [Google Scholar]
  • 5.Erbelding EJ, Post D, Stemmy E, Roberts PC, Augustine AD, Ferguson S, Paules CI, Graham BS, Fauci AS. 2018. A universal influenza vaccine: the strategic plan for the National Institute of Allergy and Infectious Diseases. J Infect Dis 218(3):347–354. doi: 10.1093/infdis/jiy103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Pearce MB, Jayaraman A, Pappas C, Belser JA, Zeng H, Gustin KM, Maines TR, Sun X, Raman R, Cox NJ, Sasisekharan R, Katz JM, Tumpey TM. 2012. Pathogenesis and transmission of swine origin A(H3N2)v influenza viruses in ferrets. Proc Natl Acad Sci U S A 109:3944–3949. doi: 10.1073/pnas.1119945109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Coughlan L, Palese P. 2018. Overcoming barriers in the path to a universal influenza virus vaccine. Cell Host Microbe 24:18–24. doi: 10.1016/j.chom.2018.06.016. [DOI] [PubMed] [Google Scholar]
  • 8.Ferdinands JM, Fry AM, Reynolds S, Petrie J, Flannery B, Jackson ML, Belongia EA. 2017. Intraseason waning of influenza vaccine protection: evidence from the US Influenza Vaccine Effectiveness Network, 2011–12 through 2014–15. Clin Infect Dis 64:544–550. doi: 10.1093/cid/ciw816. [DOI] [PubMed] [Google Scholar]
  • 9.Grohskopf LA, Sokolow LZ, Fry AM, Walter EB, Jernigan DB. 2018. Update: ACIP recommendations for the use of quadrivalent live attenuated influenza vaccine (LAIV4)—United States, 2018–19 influenza season. MMWR Morb Mortal Wkly Rep 67:643–645. doi: 10.15585/mmwr.mm6722a5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Pebody R, McMenamin J, Nohynek H. 2018. Live attenuated influenza vaccine (LAIV): recent effectiveness results from the USA and implications for LAIV programmes elsewhere. Arch Dis Child 103:101–105. doi: 10.1136/archdischild-2016-312165. [DOI] [PubMed] [Google Scholar]
  • 11.CDC. 2018. Estimated influenza illnesses, medical visits, and hospitalizations averted by vaccination in the United States. Centers for Disease Control and Prevention, Atlanta, GA: https://www.cdc.gov/flu/about/disease/2016-17.htm. Accessed 31 December 2018. [Google Scholar]
  • 12.Gomez Lorenzo MM, Fenton MJ. 2013. Immunobiology of influenza vaccines. Chest 143:502–510. doi: 10.1378/chest.12-1711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hobson D, Curry RL, Beare AS, Ward-Gardner A. 1972. The role of serum haemagglutination-inhibiting antibody in protection against challenge infection with influenza A2 and B viruses. Epidemiol Infect 70:767–777. doi: 10.1017/S0022172400022610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Potter CW, Jennings R, Nicholson K, Tyrrell DA, Dickinson KG. 1977. Immunity to attenuated influenza virus WRL 105 infection induced by heterologous, inactivated influenza A virus vaccines. J Hyg (Lond) 79:321–332. doi: 10.1017/s0022172400053158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Zost SJ, Parkhouse K, Gumina ME, Kim K, Diaz Perez S, Wilson PC, Treanor JJ, Sant AJ, Cobey S, Hensley SE. 2017. Contemporary H3N2 influenza viruses have a glycosylation site that alters binding of antibodies elicited by egg-adapted vaccine strains. Proc Natl Acad Sci U S A 114:12578–12583. doi: 10.1073/pnas.1712377114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Krammer F, Palese P. 2013. Influenza virus hemagglutinin stalk-based antibodies and vaccines. Curr Opin Virol 3:521–530. doi: 10.1016/j.coviro.2013.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kolpe A, Schepens B, Fiers W, Saelens X. 2017. M2-based influenza vaccines: recent advances and clinical potential. Expert Rev Vaccines 16:123–136. doi: 10.1080/14760584.2017.1240041. [DOI] [PubMed] [Google Scholar]
  • 18.Berthoud TK, Hamill M, Lillie PJ, Hwenda L, Collins KA, Ewer KJ, Milicic A, Poyntz HC, Lambe T, Fletcher HA, Hill AV, Gilbert SC. 2011. Potent CD8+ T-cell immunogenicity in humans of a novel heterosubtypic influenza A vaccine, MVA-NP+M1. Clin Infect Dis 52:1–7. doi: 10.1093/cid/ciq015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Campbell AJP, Grohskopf LA. 2018. Updates on influenza vaccination in children. Infect Dis Clin North Am 32:75–89. doi: 10.1016/j.idc.2017.11.005. [DOI] [PubMed] [Google Scholar]
  • 20.Neuzil KM, Jackson LA, Nelson J, Klimov A, Cox N, Bridges CB, Dunn J, DeStefano F, Shay D. 2006. Immunogenicity and reactogenicity of 1 versus 2 doses of trivalent inactivated influenza vaccine in vaccine-naive 5–8-year-old children. J Infect Dis 194:1032–1039. doi: 10.1086/507309. [DOI] [PubMed] [Google Scholar]
  • 21.Mohn KG, Smith I, Sjursen H, Cox RJ. 2018. Immune responses after live attenuated influenza vaccination. Hum Vaccin Immunother 14:571–578. doi: 10.1080/21645515.2017.1377376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lafond KE, Englund JA, Tam JS, Bresee JS. 2013. Overview of influenza vaccines in children. J Pediatric Infect Dis Soc 2:368–378. doi: 10.1093/jpids/pit053. [DOI] [PubMed] [Google Scholar]
  • 23.Osterholm MT, Kelley NS, Sommer A, Belongia EA. 2012. Efficacy and effectiveness of influenza vaccines: a systematic review and meta-analysis. Lancet Infect Dis 12:36–44. doi: 10.1016/S1473-3099(11)70295-X. [DOI] [PubMed] [Google Scholar]
  • 24.Moa AM, Chughtai AA, Muscatello DJ, Turner RM, MacIntyre CR. 2016. Immunogenicity and safety of inactivated quadrivalent influenza vaccine in adults: a systematic review and meta-analysis of randomised controlled trials. Vaccine 34:4092–4102. doi: 10.1016/j.vaccine.2016.06.064. [DOI] [PubMed] [Google Scholar]
  • 25.Goodwin K, Viboud C, Simonsen L. 2006. Antibody response to influenza vaccination in the elderly: a quantitative review. Vaccine 24:1159–1169. doi: 10.1016/j.vaccine.2005.08.105. [DOI] [PubMed] [Google Scholar]
  • 26.Belongia EA, Simpson MD, King JP, Sundaram ME, Kelley NS, Osterholm MT, McLean HQ. 2016. Variable influenza vaccine effectiveness by subtype: a systematic review and meta-analysis of test-negative design studies. Lancet Infect Dis 16:942–951. doi: 10.1016/S1473-3099(16)00129-8. [DOI] [PubMed] [Google Scholar]
  • 27.Pebody R, Warburton F, Ellis J, Andrews N, Potts A, Cottrell S, Reynolds A, Gunson R, Thompson C, Galiano M, Robertson C, Gallagher N, Sinnathamby M, Yonova I, Correa A, Moore C, Sartaj M, de Lusignan S, McMenamin J, Zambon M. 2017. End-of-season influenza vaccine effectiveness in adults and children, United Kingdom, 2016/17. Euro Surveill 22(44):17-00306. doi: 10.2807/1560-7917.ES.2017.22.44.17-00306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Rowhani-Rahbar A, Klein NP, Baxter R. 2012. Assessing the safety of influenza vaccination in specific populations: children and the elderly. Expert Rev Vaccines 11:973–984. doi: 10.1586/erv.12.66. [DOI] [PubMed] [Google Scholar]
  • 29.Mo Z, Nong Y, Liu S, Shao M, Liao X, Go K, Lavis N. 2017. Immunogenicity and safety of a trivalent inactivated influenza vaccine produced in Shenzhen, China. Hum Vaccin Immunother 13:1–7. doi: 10.1080/21645515.2017.1285475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Shen Y, Hu Y, Meng F, Du W, Li W, Song Y, Ji X, Huo L, Fu Z, Yin W. 2016. Safety, immunogenicity and cross-reactivity of a northern hemisphere 2013–2014 seasonal trivalent inactivated split influenza virus vaccine, Anflu®. Hum Vaccin Immunother 12:1229–1234. doi: 10.1080/21645515.2015.1123357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.van Duin D, Allore HG, Mohanty S, Ginter S, Newman FK, Belshe RB, Medzhitov R, Shaw AC. 2007. Prevaccine determination of the expression of costimulatory B7 molecules in activated monocytes predicts influenza vaccine responses in young and older adults. J Infect Dis 195:1590–1597. doi: 10.1086/516788. [DOI] [PubMed] [Google Scholar]
  • 32.Panda A, Qian F, Mohanty S, van Duin D, Newman FK, Zhang L, Chen S, Towle V, Belshe RB, Fikrig E, Allore HG, Montgomery RR, Shaw AC. 2010. Age-associated decrease in TLR function in primary human dendritic cells predicts influenza vaccine response. J Immunol 184:2518–2527. doi: 10.4049/jimmunol.0901022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Mohanty S, Joshi SR, Ueda I, Wilson J, Blevins TP, Siconolfi B, Meng H, Devine L, Raddassi K, Tsang S, Belshe RB, Hafler DA, Kaech SM, Kleinstein SH, Trentalange M, Allore HG, Shaw AC. 2015. Prolonged proinflammatory cytokine production in monocytes modulated by interleukin 10 after influenza vaccination in older adults. J Infect Dis 211:1174–1184. doi: 10.1093/infdis/jiu573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Nakaya HI, Hagan T, Duraisingham SS, Lee EK, Kwissa M, Rouphael N, Frasca D, Gersten M, Mehta AK, Gaujoux R, Li GM, Gupta S, Ahmed R, Mulligan MJ, Shen-Orr S, Blomberg BB, Subramaniam S, Pulendran B. 2015. Systems analysis of immunity to influenza vaccination across multiple years and in diverse populations reveals shared molecular signatures. Immunity 43:1186–1198. doi: 10.1016/j.immuni.2015.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Frasca D, Diaz A, Romero M, Phillips M, Mendez NV, Landin AM, Blomberg BB. 2012. Unique biomarkers for B-cell function predict the serum response to pandemic H1N1 influenza vaccine. Int Immunol 24:175–182. doi: 10.1093/intimm/dxr123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Frasca D, Diaz A, Romero M, Landin AM, Phillips M, Lechner SC, Ryan JG, Blomberg BB. 2010. Intrinsic defects in B cell response to seasonal influenza vaccination in elderly humans. Vaccine 28:8077–8084. doi: 10.1016/j.vaccine.2010.10.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Khurana S, Frasca D, Blomberg B, Golding H. 2012. AID activity in B cells strongly correlates with polyclonal antibody affinity maturation in-vivo following pandemic 2009-H1N1 vaccination in humans. PLoS Pathog 8:e1002920. doi: 10.1371/journal.ppat.1002920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Sasaki S, Sullivan M, Narvaez CF, Holmes TH, Furman D, Zheng NY, Nishtala M, Wrammert J, Smith K, James JA, Dekker CL, Davis MM, Wilson PC, Greenberg HB, He XS. 2011. Limited efficacy of inactivated influenza vaccine in elderly individuals is associated with decreased production of vaccine-specific antibodies. J Clin Invest 121:3109–3119. doi: 10.1172/JCI57834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Frasca D, Diaz A, Romero M, Blomberg BB. 2016. The generation of memory B cells is maintained, but the antibody response is not, in the elderly after repeated influenza immunizations. Vaccine 34:2834–2840. doi: 10.1016/j.vaccine.2016.04.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Jiang N, He J, Weinstein JA, Penland L, Sasaki S, He XS, Dekker CL, Zheng NY, Huang M, Sullivan M, Wilson PC, Greenberg HB, Davis MM, Fisher DS, Quake SR. 2013. Lineage structure of the human antibody repertoire in response to influenza vaccination. Sci Transl Med 5:171ra19. doi: 10.1126/scitranslmed.3004794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.de Bourcy CF, Angel CJ, Vollmers C, Dekker CL, Davis MM, Quake SR. 2017. Phylogenetic analysis of the human antibody repertoire reveals quantitative signatures of immune senescence and aging. Proc Natl Acad Sci U S A 114:1105–1110. doi: 10.1073/pnas.1617959114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Najarro K, Nguyen H, Chen G, Xu M, Alcorta S, Yao X, Zukley L, Metter EJ, Truong T, Lin Y, Li H, Oelke M, Xu X, Ling SM, Longo DL, Schneck J, Leng S, Ferrucci L, Weng NP. 2015. Telomere length as an indicator of the robustness of B- and T-cell response to influenza in older adults. J Infect Dis 212:1261–1269. doi: 10.1093/infdis/jiv202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Goronzy JJ, Fulbright JW, Crowson CS, Poland GA, O’Fallon WM, Weyand CM. 2001. Value of immunological markers in predicting responsiveness to influenza vaccination in elderly individuals. J Virol 75:12182–12187. doi: 10.1128/JVI.75.24.12182-12187.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Saurwein-Teissl M, Lung TL, Marx F, Gschosser C, Asch E, Blasko I, Parson W, Bock G, Schonitzer D, Trannoy E, Grubeck-Loebenstein B. 2002. Lack of antibody production following immunization in old age: association with CD8+ CD28 T cell clonal expansions and an imbalance in the production of Th1 and Th2 cytokines. J Immunol 168:5893–5899. doi: 10.4049/jimmunol.168.11.5893. [DOI] [PubMed] [Google Scholar]
  • 45.Kang I, Hong MS, Nolasco H, Park SH, Dan JM, Choi JY, Craft J. 2004. Age-associated change in the frequency of memory CD4+ T cells impairs long term CD4+ T cell responses to influenza vaccine. J Immunol 173:673–681. doi: 10.4049/jimmunol.173.1.673. [DOI] [PubMed] [Google Scholar]
  • 46.Herati RS, Reuter MA, Dolfi DV, Mansfield KD, Aung H, Badwan OZ, Kurupati RK, Kannan S, Ertl H, Schmader KE, Betts MR, Canaday DH, Wherry EJ. 2014. Circulating CXCR5+ PD-1+ response predicts influenza vaccine antibody responses in young adults but not elderly adults. J Immunol 193:3528–3537. doi: 10.4049/jimmunol.1302503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.McElhaney JE, Ewen C, Zhou X, Kane KP, Xie D, Hager WD, Barry MB, Kleppinger A, Wang Y, Bleackley RC. 2009. Granzyme B: correlates with protection and enhanced CTL response to influenza vaccination in older adults. Vaccine 27:2418–2425. doi: 10.1016/j.vaccine.2009.01.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Zhou X, McElhaney JE. 2011. Age-related changes in memory and effector T cells responding to influenza A/H3N2 and pandemic A/H1N1 strains in humans. Vaccine 29:2169–2177. doi: 10.1016/j.vaccine.2010.12.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Wagar LE, Gentleman B, Pircher H, McElhaney JE, Watts TH. 2011. Influenza-specific T cells from older people are enriched in the late effector subset and their presence inversely correlates with vaccine response. PLoS One 6:e23698. doi: 10.1371/journal.pone.0023698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Merani S, Pawelec G, Kuchel GA, McElhaney JE. 2017. Impact of aging and cytomegalovirus on immunological response to influenza vaccination and infection. Front Immunol 8:784. doi: 10.3389/fimmu.2017.00784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Henry C, Zheng NY, Huang M, Cabanov A, Rojas KT, Kaur K, Andrews SF, Palm AE, Chen YQ, Li Y, Hoskova K, Utset HA, Vieira MC, Wrammert J, Ahmed R, Holden-Wiltse J, Topham DJ, Treanor JJ, Ertl HC, Schmader KE, Cobey S, Krammer F, Hensley SE, Greenberg H, He XS, Wilson PC. 2019. Influenza virus vaccination elicits poorly adapted B cell responses in elderly individuals. Cell Host Microbe 25(3):P357–366.E6. doi: 10.1016/j.chom.2019.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.DiazGranados CA, Dunning AJ, Kimmel M, Kirby D, Treanor J, Collins A, Pollak R, Christoff J, Earl J, Landolfi V, Martin E, Gurunathan S, Nathan R, Greenberg DP, Tornieporth NG, Decker MD, Talbot HK. 2014. Efficacy of high-dose versus standard-dose influenza vaccine in older adults. N Engl J Med 371:635–645. doi: 10.1056/NEJMoa1315727. [DOI] [PubMed] [Google Scholar]
  • 53.Van Buynder PG, Konrad S, Van Buynder JL, Brodkin E, Krajden M, Ramler G, Bigham M. 2013. The comparative effectiveness of adjuvanted and unadjuvanted trivalent inactivated influenza vaccine (TIV) in the elderly. Vaccine 31:6122–6128. doi: 10.1016/j.vaccine.2013.07.059. [DOI] [PubMed] [Google Scholar]
  • 54.McDonald JU, Zhong Z, Groves HT, Tregoning JS. 2017. Inflammatory responses to influenza vaccination at the extremes of age. Immunology 151:451–463. doi: 10.1111/imm.12742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Ramirez A, Co M, Mathew A. 2016. CpG improves influenza vaccine efficacy in young adult but not aged mice. PLoS One 11:e0150425. doi: 10.1371/journal.pone.0150425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Potluri T, Fink AL, Sylvia KE, Dhakal S, Vermillion MS, Vom Steeg L, Deshpande S, Narasimhan H, Klein SL. 2019. Age-associated changes in the impact of sex steroids on influenza vaccine responses in males and females. NPJ Vaccines 4:29. doi: 10.1038/s41541-019-0124-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Dawany N, Parzych EM, Showe LC, Ertl HC. 2016. Age-related changes in the gene expression profile of antigen-specific mouse CD8+ T cells can be partially reversed by blockade of the BTLA/CD160 pathways during vaccination. Aging (Albany NY) 8:3272–3297. doi: 10.18632/aging.101105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lefebvre JS, Lorenzo EC, Masters AR, Hopkins JW, Eaton SM, Smiley ST, Haynes L. 2016. Vaccine efficacy and T helper cell differentiation change with aging. Oncotarget 7:33581–33594. doi: 10.18632/oncotarget.9254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Baldwin SL, Hsu FC, Van Hoeven N, Gage E, Granger B, Guderian JA, Larsen SE, Lorenzo EC, Haynes L, Reed SG, Coler RN. 2018. Improved immune responses in young and aged mice with adjuvanted vaccines against H1N1 influenza infection. Front Immunol 9:295. doi: 10.3389/fimmu.2018.00295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Yam KK, Gupta J, Allen EK, Burt KR, Beaulieu E, Mallett CP, Burt DS, Ward BJ. 2016. Comparison of AS03 and alum on immune responses elicited by A/H3N2 split influenza vaccine in young, mature and aged BALB/c mice. Vaccine 34:1444–1451. doi: 10.1016/j.vaccine.2016.02.012. [DOI] [PubMed] [Google Scholar]
  • 61.Garcia M, Misplon JA, Price GE, Lo CY, Epstein SL. 2016. Age dependence of immunity induced by a candidate universal influenza vaccine in mice. PLoS One 11:e0153195. doi: 10.1371/journal.pone.0153195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Flanagan KL, Fink AL, Plebanski M, Klein SL. 2017. Sex and gender differences in the outcomes of vaccination over the life course. Annu Rev Cell Dev Biol 33:577–599. doi: 10.1146/annurev-cellbio-100616-060718. [DOI] [PubMed] [Google Scholar]
  • 63.Engler RJ, Nelson MR, Klote MM, VanRaden MJ, Huang CY, Cox NJ, Klimov A, Keitel WA, Nichol KL, Carr WW, Treanor JJ, Walter Reed Health Care System Influenza Vaccine Center. 2008. Half- vs. full-dose trivalent inactivated influenza vaccine (2004–2005): age, dose, and sex effects on immune responses. Arch Intern Med 168:2405–2414. doi: 10.1001/archinternmed.2008.513. [DOI] [PubMed] [Google Scholar]
  • 64.Furman D, Hejblum BP, Simon N, Jojic V, Dekker CL, Thiebaut R, Tibshirani RJ, Davis MM. 2014. Systems analysis of sex differences reveals an immunosuppressive role for testosterone in the response to influenza vaccination. Proc Natl Acad Sci U S A 111:869–874. doi: 10.1073/pnas.1321060111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Cook IF, Barr I, Hartel G, Pond D, Hampson AW. 2006. Reactogenicity and immunogenicity of an inactivated influenza vaccine administered by intramuscular or subcutaneous injection in elderly adults. Vaccine 24:2395–2402. doi: 10.1016/j.vaccine.2005.11.057. [DOI] [PubMed] [Google Scholar]
  • 66.Falsey AR, Treanor JJ, Tornieporth N, Capellan J, Gorse GJ. 2009. 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. J Infect Dis 200:172–180. doi: 10.1086/599790. [DOI] [PubMed] [Google Scholar]
  • 67.Kao TM, Hsieh SM, Kung HC, Lee YC, Huang KC, Huang LM, Chang FY, Wang NC, Liu YC, Lee WS, Liu HE, Chen CI, Chen CH. 2010. Immune response of single dose vaccination against 2009 pandemic influenza A (H1N1) in the Taiwanese elderly. Vaccine 28:6159–6163. doi: 10.1016/j.vaccine.2010.07.026. [DOI] [PubMed] [Google Scholar]
  • 68.Talaat KR, Greenberg ME, Lai MH, Hartel GF, Wichems CH, Rockman S, Jeanfreau RJ, Ghosh MR, Kabongo ML, Gittleson C, Karron RA. 2010. A single dose of unadjuvanted novel 2009 H1N1 vaccine is immunogenic and well tolerated in young and elderly adults. J Infect Dis 202:1327–1337. doi: 10.1086/656601. [DOI] [PubMed] [Google Scholar]
  • 69.Fleming DM, Watson JM, Nicholas S, Smith GE, Swan AV. 1995. Study of the effectiveness of influenza vaccination in the elderly in the epidemic of 1989–90 using a general practice database. Epidemiol Infect 115:581–589. doi: 10.1017/s095026880005874x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wang CS, Wang ST, Chou P. 2002. Efficacy and cost-effectiveness of influenza vaccination of the elderly in a densely populated and unvaccinated community. Vaccine 20:2494–2499. doi: 10.1016/S0264-410X(02)00181-0. [DOI] [PubMed] [Google Scholar]
  • 71.Vila-Corcoles A, Rodriguez T, de Diego C, Ochoa O, Valdivieso A, Salsench E, Ansa X, Badia W, Saun N, Group ES. 2007. Effect of influenza vaccine status on winter mortality in Spanish community-dwelling elderly people during 2002–2005 influenza periods. Vaccine 25:6699–6707. doi: 10.1016/j.vaccine.2007.07.015. [DOI] [PubMed] [Google Scholar]
  • 72.Chambers C, Skowronski DM, Rose C, Serres G, Winter AL, Dickinson JA, Jassem A, Gubbay JB, Fonseca K, Drews SJ, Charest H, Martineau C, Petric M, Krajden M. 2018. Should sex be considered an effect modifier in the evaluation of influenza vaccine effectiveness? Open Forum Infect Dis 5:ofy211. doi: 10.1093/ofid/ofy211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lorenzo ME, Hodgson A, Robinson DP, Kaplan JB, Pekosz A, Klein SL. 2011. Antibody responses and cross protection against lethal influenza A viruses differ between the sexes in C57BL/6 mice. Vaccine 29:9246–9255. doi: 10.1016/j.vaccine.2011.09.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Živković I, Bufan B, Petrušić V, Minić R, Arsenović-Ranin N, Petrović R, Leposavić G. 2015. Sexual diergism in antibody response to whole virus trivalent inactivated influenza vaccine in outbred mice. Vaccine 33:5546–5552. doi: 10.1016/j.vaccine.2015.09.006. [DOI] [PubMed] [Google Scholar]
  • 75.Živković I, Petrović R, Arsenović-Ranin N, Petrušić V, Minić R, Bufan B, Popović O, Leposavić G. 2018. Sex bias in mouse humoral immune response to influenza vaccine depends on the vaccine type. Biologicals 52:18–24. doi: 10.1016/j.biologicals.2018.01.007. [DOI] [PubMed] [Google Scholar]
  • 76.Fink AL, Engle K, Ursin RL, Tang WY, Klein SL. 2018. Biological sex affects vaccine efficacy and protection against influenza in mice. Proc Natl Acad Sci U S A doi: 10.1073/pnas.1805268115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Pone EJ, Lou Z, Lam T, Greenberg ML, Wang R, Xu Z, Casali P. 2015. B cell TLR1/2, TLR4, TLR7 and TLR9 interact in induction of class switch DNA recombination: modulation by BCR and CD40, and relevance to T-independent antibody responses. Autoimmunity 48:1–12. doi: 10.3109/08916934.2014.993027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Fan H, Dong G, Zhao G, Liu F, Yao G, Zhu Y, Hou Y. 2014. Gender differences of B cell signature in healthy subjects underlie disparities in incidence and course of SLE related to estrogen. J Immunol Res 2014:814598. doi: 10.1155/2014/814598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Nguyen DC, Masseoud F, Lu X, Scinicariello F, Sambhara S, Attanasio R. 2011. 17beta-Estradiol restores antibody responses to an influenza vaccine in a postmenopausal mouse model. Vaccine 29:2515–2518. doi: 10.1016/j.vaccine.2011.01.080. [DOI] [PubMed] [Google Scholar]
  • 80.Henry C, Palm AE, Krammer F, Wilson PC. 2018. From original antigenic sin to the universal influenza virus vaccine. Trends Immunol 39:70–79. doi: 10.1016/j.it.2017.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Bodewes R, de Mutsert G, van der Klis FR, Ventresca M, Wilks S, Smith DJ, Koopmans M, Fouchier RA, Osterhaus AD, Rimmelzwaan GF. 2011. Prevalence of antibodies against seasonal influenza A and B viruses in children in Netherlands. Clin Vaccine Immunol 18:469–476. doi: 10.1128/CVI.00396-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Francis T., Jr 1960. On the doctrine of original antigenic sin. Proc Am Philos Soc 104:572–578. [Google Scholar]
  • 83.Lessler J, Riley S, Read JM, Wang S, Zhu H, Smith GJ, Guan Y, Jiang CQ, Cummings DA. 2012. Evidence for antigenic seniority in influenza A (H3N2) antibody responses in southern China. PLoS Pathog 8:e1002802. doi: 10.1371/journal.ppat.1002802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Miller MS, Gardner TJ, Krammer F, Aguado LC, Tortorella D, Basler CF, Palese P. 2013. Neutralizing antibodies against previously encountered influenza virus strains increase over time: a longitudinal analysis. Sci Transl Med 5:198ra107. doi: 10.1126/scitranslmed.3006637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Cortina-Ceballos B, Godoy-Lozano EE, Téllez-Sosa J, Ovilla-Muñoz M, Sámano-Sánchez H, Aguilar-Salgado A, Gómez-Barreto RE, Valdovinos-Torres H, López-Martínez I, Aparicio-Antonio R, Rodríguez MH, Martínez-Barnetche J. 2015. Longitudinal analysis of the peripheral B cell repertoire reveals unique effects of immunization with a new influenza virus strain. Genome Med 7:124. doi: 10.1186/s13073-015-0239-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Tan YC, Blum LK, Kongpachith S, Ju CH, Cai X, Lindstrom TM, Sokolove J, Robinson WH. 2014. High-throughput sequencing of natively paired antibody chains provides evidence for original antigenic sin shaping the antibody response to influenza vaccination. Clin Immunol 151:55–65. doi: 10.1016/j.clim.2013.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Webster RG. 1966. Original antigenic sin in ferrets: the response to sequential infections with influenza viruses. J Immunol 97:177–183. [PubMed] [Google Scholar]
  • 88.Angelova LA, Shvartsman Ya S. 1982. Original antigenic sin to influenza in rats. Immunology 46:183–188. [PMC free article] [PubMed] [Google Scholar]
  • 89.Kim JH, Liepkalns J, Reber AJ, Lu X, Music N, Jacob J, Sambhara S. 2016. Prior infection with influenza virus but not vaccination leaves a long-term immunological imprint that intensifies the protective efficacy of antigenically drifted vaccine strains. Vaccine 34:495–502. doi: 10.1016/j.vaccine.2015.11.077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Zhang A, Stacey HD, Mullarkey CE, Miller MS. 2019. Original antigenic sin: how first exposure shapes lifelong anti-influenza virus immune responses. J Immunol 202:335–340. doi: 10.4049/jimmunol.1801149. [DOI] [PubMed] [Google Scholar]
  • 91.Guthmiller JJ, Wilson PC. 2018. Harnessing immune history to combat influenza viruses. Curr Opin Immunol 53:187–195. doi: 10.1016/j.coi.2018.05.010. [DOI] [PubMed] [Google Scholar]
  • 92.Linderman SL, Chambers BS, Zost SJ, Parkhouse K, Li Y, Herrmann C, Ellebedy AH, Carter DM, Andrews SF, Zheng NY, Huang M, Huang Y, Strauss D, Shaz BH, Hodinka RL, Reyes-Teran G, Ross TM, Wilson PC, Ahmed R, Bloom JD, Hensley SE. 2014. Potential antigenic explanation for atypical H1N1 infections among middle-aged adults during the 2013–2014 influenza season. Proc Natl Acad Sci U S A 111:15798–15803. doi: 10.1073/pnas.1409171111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Flannery B, Smith C, Garten RJ, Levine MZ, Chung JR, Jackson ML, Jackson LA, Monto AS, Martin ET, Belongia EA, McLean HQ, Gaglani M, Murthy K, Zimmerman R, Nowalk MP, Griffin MR, Keipp Talbot H, Treanor JJ, Wentworth DE, Fry AM. 2018. Influence of birth cohort on effectiveness of 2015–2016 influenza vaccine against medically attended illness due to 2009 pandemic influenza A(H1N1) virus in the United States. J Infect Dis 218:189–196. doi: 10.1093/infdis/jix634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Skowronski DM, Chambers C, Sabaiduc S, De Serres G, Winter AL, Dickinson JA, Gubbay JB, Drews SJ, Martineau C, Charest H, Krajden M, Bastien N, Li Y. 2017. Beyond antigenic match: possible agent-host and immuno-epidemiological influences on influenza vaccine effectiveness during the 2015–2016 season in Canada. J Infect Dis 216:1487–1500. doi: 10.1093/infdis/jix526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Kim JH, Davis WG, Sambhara S, Jacob J. 2012. Strategies to alleviate original antigenic sin responses to influenza viruses. Proc Natl Acad Sci U S A 109:13751–13756. doi: 10.1073/pnas.0912458109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Domnich A, Arata L, Amicizia D, Puig-Barbera J, Gasparini R, Panatto D. 2017. Effectiveness of MF59-adjuvanted seasonal influenza vaccine in the elderly: a systematic review and meta-analysis. Vaccine 35:513–520. doi: 10.1016/j.vaccine.2016.12.011. [DOI] [PubMed] [Google Scholar]
  • 97.Xu R, Ekiert DC, Krause JC, Hai R, Crowe JE Jr, Wilson IA. 2010. Structural basis of preexisting immunity to the 2009 H1N1 pandemic influenza virus. Science 328:357–360. doi: 10.1126/science.1186430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Worobey M, Han GZ, Rambaut A. 2014. Genesis and pathogenesis of the 1918 pandemic H1N1 influenza A virus. Proc Natl Acad Sci U S A 111:8107–8112. doi: 10.1073/pnas.1324197111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Gostic KM, Ambrose M, Worobey M, Lloyd-Smith JO. 2016. Potent protection against H5N1 and H7N9 influenza via childhood hemagglutinin imprinting. Science 354:722–726. doi: 10.1126/science.aag1322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Morelli SS, Mandal M, Goldsmith LT, Kashani BN, Ponzio NM. 2015. The maternal immune system during pregnancy and its influence on fetal development. Res Rep Biol 6:171–189. doi: 10.2147/RRB.S80652. [DOI] [Google Scholar]
  • 101.Aghaeepour N, Ganio EA, McIlwain D, Tsai AS, Tingle M, Van Gassen S, Gaudilliere DK, Baca Q, McNeil L, Okada R, Ghaemi MS, Furman D, Wong RJ, Winn VD, Druzin ML, El-Sayed YY, Quaintance C, Gibbs R, Darmstadt GL, Shaw GM, Stevenson DK, Tibshirani R, Nolan GP, Lewis DB, Angst MS, Gaudilliere B. 2017. An immune clock of human pregnancy. Sci Immunol 2(15):eaan2946. doi: 10.1126/sciimmunol.aan2946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Mertz D, Geraci J, Winkup J, Gessner BD, Ortiz JR, Loeb M. 2017. Pregnancy as a risk factor for severe outcomes from influenza virus infection: a systematic review and meta-analysis of observational studies. Vaccine 35:521–528. doi: 10.1016/j.vaccine.2016.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Sullivan SG, Price OH, Regan AK. 2019. Burden, effectiveness and safety of influenza vaccines in elderly, paediatric and pregnant populations. Ther Adv Vaccines Immunother 7:2515135519826481. doi: 10.1177/2515135519826481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Sperling RS, Engel SM, Wallenstein S, Kraus TA, Garrido J, Singh T, Kellerman L, Moran TM. 2012. Immunogenicity of trivalent inactivated influenza vaccination received during pregnancy or postpartum. Obstet Gynecol 119:631–639. doi: 10.1097/AOG.0b013e318244ed20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Kay AW, Bayless NL, Fukuyama J, Aziz N, Dekker CL, Mackey S, Swan GE, Davis MM, Blish CA. 2015. Pregnancy does not attenuate the antibody or plasmablast response to inactivated influenza vaccine. J Infect Dis 212:861–870. doi: 10.1093/infdis/jiv138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Murray DL, Imagawa DT, Okada DM, St Geme JW Jr. 1979. Antibody response to monovalent A/New Jersey/8/76 influenza vaccine in pregnant women. J Clin Microbiol 10:184–187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Steinhoff MC, Katz J, Englund JA, Khatry SK, Shrestha L, Kuypers J, Stewart L, Mullany LC, Chu HY, LeClerq SC, Kozuki N, McNeal M, Reedy AM, Tielsch JM. 2017. Year-round influenza immunization during pregnancy in Nepal: a phase 4, randomised, placebo-controlled trial. Lancet Infect Dis 17:981–989. doi: 10.1016/S1473-3099(17)30252-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Mutsaerts E, Madhi SA, Cutland CL, Jones S, Hugo A, Trenor S, Treurnicht FK, Klipstein-Grobusch K, Weinberg A, Nunes MC. 2016. Influenza vaccination of pregnant women protects them over two consecutive influenza seasons in a randomized controlled trial. Expert Rev Vaccines 15:1055–1062. doi: 10.1080/14760584.2016.1192473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Zaman K, Roy E, Arifeen SE, Rahman M, Raqib R, Wilson E, Omer SB, Shahid NS, Breiman RF, Breiman RE, Steinhoff MC. 2008. Effectiveness of maternal influenza immunization in mothers and infants. N Engl J Med 359:1555–1564. doi: 10.1056/NEJMoa0708630. [DOI] [PubMed] [Google Scholar]
  • 110.Moro P, Baumblatt J, Lewis P, Cragan J, Tepper N, Cano M. 2017. Surveillance of adverse events after seasonal influenza vaccination in pregnant women and their infants in the vaccine adverse event reporting system, July 2010–May 2016. Drug Saf 40:145–152. doi: 10.1007/s40264-016-0482-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Kharbanda EO, Vazquez-Benitez G, Romitti PA, Naleway AL, Cheetham TC, Lipkind HS, Klein NP, Lee G, Jackson ML, Hambidge SJ, McCarthy N, DeStefano F, Nordin JD, Vaccine Safety D. 2017. First trimester influenza vaccination and risks for major structural birth defects in offspring. J Pediatr 187:234–239.e4. doi: 10.1016/j.jpeds.2017.04.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Donahue JG, Kieke BA, King JP, DeStefano F, Mascola MA, Irving SA, Cheetham TC, Glanz JM, Jackson LA, Klein NP, Naleway AL, Weintraub E, Belongia EA. 2017. Association of spontaneous abortion with receipt of inactivated influenza vaccine containing H1N1pdm09 in 2010–11 and 2011–12. Vaccine 35:5314–5322. doi: 10.1016/j.vaccine.2017.06.069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Eke AC, Dooley KE, Sheffield JS. 2019. Pharmacologic research in pregnant women: time to get it right. N Engl J Med 380:1293–1295. doi: 10.1056/NEJMp1815325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Marcelin G, Aldridge JR, Duan S, Ghoneim HE, Rehg J, Marjuki H, Boon AC, McCullers JA, Webby RJ. 2011. Fatal outcome of pandemic H1N1 2009 influenza virus infection is associated with immunopathology and impaired lung repair, not enhanced viral burden, in pregnant mice. J Virol 85:11208–11219. doi: 10.1128/JVI.00654-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Littauer EQ, Esser ES, Antao OQ, Vassilieva EV, Compans RW, Skountzou I. 2017. H1N1 influenza virus infection results in adverse pregnancy outcomes by disrupting tissue-specific hormonal regulation. PLoS Pathog 13:e1006757. doi: 10.1371/journal.ppat.1006757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Mbawuike IN, Six HR, Cate TR, Couch RB. 1990. Vaccination with inactivated influenza A virus during pregnancy protects neonatal mice against lethal challenge by influenza A viruses representing three subtypes. J Virol 64:1370–1374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Hwang SD, Shin JS, Ku KB, Kim HS, Cho SW, Seo SH. 2010. Protection of pregnant mice, fetuses and neonates from lethality of H5N1 influenza viruses by maternal vaccination. Vaccine 28:2957–2964. doi: 10.1016/j.vaccine.2010.02.016. [DOI] [PubMed] [Google Scholar]
  • 118.Louie JK, Acosta M, Winter K, Jean C, Gavali S, Schechter R, Vugia D, Harriman K, Matyas B, Glaser CA, Samuel MC, Rosenberg J, Talarico J, Hatch D, California Pandemic Working G. 2009. Factors associated with death or hospitalization due to pandemic 2009 influenza A(H1N1) infection in California. JAMA 302:1896–1902. doi: 10.1001/jama.2009.1583. [DOI] [PubMed] [Google Scholar]
  • 119.Neidich SD, Green WD, Rebeles J, Karlsson EA, Schultz-Cherry S, Noah TL, Chakladar S, Hudgens MG, Weir SS, Beck MA. 2017. Increased risk of influenza among vaccinated adults who are obese. Int J Obes 41:1324–1330. doi: 10.1038/ijo.2017.131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Sheridan PA, Paich HA, Handy J, Karlsson EA, Hudgens MG, Sammon AB, Holland LA, Weir S, Noah TL, Beck MA. 2012. Obesity is associated with impaired immune response to influenza vaccination in humans. Int J Obes 36:1072–1077. doi: 10.1038/ijo.2011.208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Smit MA, Wang H-L, Kim E, Barragan N, Aldrovandi GM, El Amin AN, Mascola L, Pannaraj PS. 2016. Influenza vaccine is protective against laboratory-confirmed influenza in obese children. Pediatr Infect Dis J 35:440–445. doi: 10.1097/INF.0000000000001029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Park HL, Shim SH, Lee EY, Cho W, Park S, Jeon HJ, Ahn SY, Kim H, Nam JH. 2014. Obesity-induced chronic inflammation is associated with the reduced efficacy of influenza vaccine. Hum Vaccin Immunother 10:1181–1186. doi: 10.4161/hv.28332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Karlsson EA, Hertz T, Johnson C, Mehle A, Krammer F, Schultz-Cherry S. 2016. Obesity outweighs protection conferred by adjuvanted influenza vaccination. mBio 7(4):e01144-16. doi: 10.1128/mBio.01144-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Castaner O, Goday A, Park YM, Lee SH, Magkos F, Shiow STE, Schroder H. 2018. The gut microbiome profile in obesity: a systematic review. Int J Endocrinol 2018:4095789. doi: 10.1155/2018/4095789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Belkaid Y, Hand TW. 2014. Role of the microbiota in immunity and inflammation. Cell 157:121–141. doi: 10.1016/j.cell.2014.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Abt MC, Osborne LC, Monticelli LA, Doering TA, Alenghat T, Sonnenberg GF, Paley MA, Antenus M, Williams KL, Erikson J, Wherry EJ, Artis D. 2012. Commensal bacteria calibrate the activation threshold of innate antiviral immunity. Immunity 37:158–170. doi: 10.1016/j.immuni.2012.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Ichinohe T, Pang IK, Kumamoto Y, Peaper DR, Ho JH, Murray TS, Iwasaki A. 2011. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc Natl Acad Sci U S A 108:5354–5359. doi: 10.1073/pnas.1019378108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Wu S, Jiang ZY, Sun YF, Yu B, Chen J, Dai CQ, Wu XL, Tang XL, Chen XY. 2013. Microbiota regulates the TLR7 signaling pathway against respiratory tract influenza A virus infection. Curr Microbiol 67:414–422. doi: 10.1007/s00284-013-0380-z. [DOI] [PubMed] [Google Scholar]
  • 129.Youn HN, Lee DH, Lee YN, Park JK, Yuk SS, Yang SY, Lee HJ, Woo SH, Kim HM, Lee JB, Park SY, Choi IS, Song CS. 2012. Intranasal administration of live Lactobacillus species facilitates protection against influenza virus infection in mice. Antiviral Res 93:138–143. doi: 10.1016/j.antiviral.2011.11.004. [DOI] [PubMed] [Google Scholar]
  • 130.Asama T, Uematsu T, Kobayashi N, Tatefuji T, Hashimoto K. 2017. Oral administration of heat-killed Lactobacillus kunkeei YB38 improves murine influenza pneumonia by enhancing IgA production. Biosci Microbiota Food Health 36:1–9. doi: 10.12938/bmfh.16-010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Shi SH, Yang WT, Yang GL, Zhang XK, Liu YY, Zhang LJ, Ye LP, Hu JT, Xing X, Qi C, Li Y, Wang CF. 2016. Lactobacillus plantarum vaccine vector expressing hemagglutinin provides protection against H9N2 challenge infection. Virus Res 211:46–57. doi: 10.1016/j.virusres.2015.09.005. [DOI] [PubMed] [Google Scholar]
  • 132.Yang WT, Yang GL, Wang Q, Huang HB, Jiang YL, Shi CW, Wang JZ, Huang KY, Jin YB, Wang CF. 2017. Protective efficacy of Fc targeting conserved influenza virus M2e antigen expressed by Lactobacillus plantarum. Antiviral Res 138:9–21. doi: 10.1016/j.antiviral.2016.11.025. [DOI] [PubMed] [Google Scholar]
  • 133.Rizzardini G, Eskesen D, Calder PC, Capetti A, Jespersen L, Clerici M. 2012. Evaluation of the immune benefits of two probiotic strains Bifidobacterium animalis ssp. lactis, BB-12(R) and Lactobacillus paracasei ssp. paracasei, L. casei 431(R) in an influenza vaccination model: a randomised, double-blind, placebo-controlled study. Br J Nutr 107:876–884. doi: 10.1017/S000711451100420X. [DOI] [PubMed] [Google Scholar]
  • 134.Davidson LE, Fiorino AM, Snydman DR, Hibberd PL. 2011. Lactobacillus GG as an immune adjuvant for live-attenuated influenza vaccine in healthy adults: a randomized double-blind placebo-controlled trial. Eur J Clin Nutr 65:501–507. doi: 10.1038/ejcn.2010.289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Bosch M, Mendez M, Perez M, Farran A, Fuentes MC, Cune J. 2012. Lactobacillus plantarum CECT7315 and CECT7316 stimulate immunoglobulin production after influenza vaccination in elderly. Nutr Hosp 27:504–509. [DOI] [PubMed] [Google Scholar]
  • 136.Akatsu H, Arakawa K, Yamamoto T, Kanematsu T, Matsukawa N, Ohara H, Maruyama M. 2013. Lactobacillus in jelly enhances the effect of influenza vaccination in elderly individuals. J Am Geriatr Soc 61:1828–1830. doi: 10.1111/jgs.12474. [DOI] [PubMed] [Google Scholar]
  • 137.Nakaya HI, Wrammert J, Lee EK, Racioppi L, Marie-Kunze S, Haining WN, Means AR, Kasturi SP, Khan N, Li GM, McCausland M, Kanchan V, Kokko KE, Li S, Elbein R, Mehta AK, Aderem A, Subbarao K, Ahmed R, Pulendran B. 2011. Systems biology of vaccination for seasonal influenza in humans. Nat Immunol 12:786–795. doi: 10.1038/ni.2067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.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 41:478–492. doi: 10.1016/j.immuni.2014.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Poland GA, Ovsyannikova IG, Jacobson RM. 2008. Immunogenetics of seasonal influenza vaccine response. Vaccine 26(Suppl 4):D35–D40. doi: 10.1016/j.vaccine.2008.07.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Chen T, Xiao M, Yang J, Chen YK, Bai T, Tang XJ, Shu YL. 2018. Association between rs12252 and influenza susceptibility and severity: an updated meta-analysis. Epidemiol Infect doi: 10.1017/S0950268818002832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Prabhu SS, Chakraborty TT, Kumar N, Banerjee I. 2018. Association between IFITM3 rs12252 polymorphism and influenza susceptibility and severity: a meta-analysis. Gene 674:70–79. doi: 10.1016/j.gene.2018.06.070. [DOI] [PubMed] [Google Scholar]
  • 142.Bailey CC, Zhong G, Huang IC, Farzan M. 2014. IFITM-family proteins: the cell’s first line of antiviral defense. Annu Rev Virol 1:261–283. doi: 10.1146/annurev-virology-031413-085537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Franco LM, Bucasas KL, Wells JM, Nino D, Wang X, Zapata GE, Arden N, Renwick A, Yu P, Quarles JM, Bray MS, Couch RB, Belmont JW, Shaw CA. 2013. Integrative genomic analysis of the human immune response to influenza vaccination. Elife 2:e00299. doi: 10.7554/eLife.00299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Moss AJ, Gaughran FP, Karasu A, Gilbert AS, Mann AJ, Gelder CM, Oxford JS, Stephens HA, Lambkin-Williams R. 2013. Correlation between human leukocyte antigen class II alleles and HAI titers detected post-influenza vaccination. PLoS One 8:e71376. doi: 10.1371/journal.pone.0071376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Avnir Y, Watson CT, Glanville J, Peterson EC, Tallarico AS, Bennett AS, Qin K, Fu Y, Huang CY, Beigel JH, Breden F, Zhu Q, Marasco WA. 2016. IGHV1-69 polymorphism modulates anti-influenza antibody repertoires, correlates with IGHV utilization shifts and varies by ethnicity. Sci Rep 6:20842. doi: 10.1038/srep20842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Vom Steeg LG, Klein SL. 2019. Sex and sex steroids impact influenza pathogenesis across the life course. Semin Immunopathol 41:189–194. doi: 10.1007/s00281-018-0718-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Pauklin S, Sernandez IV, Bachmann G, Ramiro AR, Petersen-Mahrt SK. 2009. Estrogen directly activates AID transcription and function. J Exp Med 206:99–111. doi: 10.1084/jem.20080521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Deguchi K, Kamada M, Irahara M, Maegawa M, Yamamoto S, Ohmoto Y, Murata K, Yasui T, Yamano S, Aono T. 2001. Postmenopausal changes in production of type 1 and type 2 cytokines and the effects of hormone replacement therapy. Menopause 8:266–273. doi: 10.1097/00042192-200107000-00008. [DOI] [PubMed] [Google Scholar]
  • 149.Kamada M, Irahara M, Maegawa M, Yasui T, Yamano S, Yamada M, Tezuka M, Kasai Y, Deguchi K, Ohmoto Y, Aono T. 2001. B cell subsets in postmenopausal women and the effect of hormone replacement therapy. Maturitas 37:173–179. doi: 10.1016/S0378-5122(00)00180-8. [DOI] [PubMed] [Google Scholar]
  • 150.Engelmann F, Rivera A, Park B, Messerle-Forbes M, Jensen JT, Messaoudi I. 2016. Impact of estrogen therapy on lymphocyte homeostasis and the response to seasonal influenza vaccine in post-menopausal women. PLoS One 11:e0149045. doi: 10.1371/journal.pone.0149045. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Virology are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES