1,597
Views
11
CrossRef citations to date
0
Altmetric
Review

Factors influencing the immunogenicity of influenza vaccines

ORCID Icon, , , &
Pages 2706-2718 | Received 18 Oct 2020, Accepted 07 Jan 2021, Published online: 11 Mar 2021

ABSTRACT

Annual vaccination is the best prevention of influenza. However, the immunogenicity of influenza vaccines varies among different populations. It is important to fully identify the factors that may affect the immunogenicity of the vaccines to provide best protection for vaccine recipients. This paper reviews the factors that may influence the immunogenicity of influenza vaccines from the aspects of vaccine factors, adjuvants, individual factors, repeated vaccination, and genetic factors. The confirmed or hypothesized molecular mechanisms of these factors have also been briefly summarized.

Introduction

Influenza is an acute infectious respiratory disease, which is mainly transmitted by droplets and human-to-human close contacts. It is estimated that there are 1 billion cases worldwide annually, including 3–5 million serious cases and 290,000–650,000 deaths.Citation1

The most effective measure to prevent influenza infection is by taking annual vaccination. However, for some special populations, including pregnant women, infants, elderly people and chronic patients, influenza still threats their life and health due to the poor effectiveness of current influenza vaccines. Pregnancy confers an increased risk for hospitalization with influenza. It was estimated that the risk of influenza-associated hospitalization in pregnant women was significantly higher than nonpregnant ones, with an odds ratio of 2.44.Citation2,Citation3 WHO recommended that pregnant women should be the prior population for influenza vaccination. For infants, a variety of complications such as pneumonia and myocarditis can be triggered after influenza infection, some studies also reported that the infection can even cause encephalopathy and encephalitis among infants.Citation4–6 When elderly people are infected with influenza, their conditions are usually developed rapidly and may even cause death.Citation7 For people with other basic diseases, the infection with influenza viruses will not only lead to severe conditions, sometimes will also aggravate their original diseases. Thus, in order to provide a better protection for these people, it is of great significance to identify the correlative factors that may influence the influenza vaccine effectiveness (VE).

There are mainly two factors that may influence the VE. One is the matching degree of circulating strains and vaccine strains, and the other is the immunogenicity of the vaccine. Many studies have confirmed that the estimates of VE vary widely from season to season. And the VE will decrease when the vaccine strains and circulating strains are poorly matched.Citation8,Citation9 A meta-analysis study showed that in poorly matched seasons, the risk ratio (RR) of being hospitalized for influenza was 2.04 (95%CI = 1.29–3.22), whereas in well-matched seasons, the RR was only 0.64 (95%CI = 0.33–1.22), suggesting the importance of effective prediction for annual influenza vaccine strains.Citation10 On the other hand, the immunogenicity of the vaccine, which means, the vaccine’s capacity of inducing immune response after inoculation, also has an important impact on the VE. The immunogenicity of influenza vaccine mainly can be evaluated by three indicators: the geometric mean titer (GMT) of antibodies, the seroprotection rate, and the seroconversion rate. Many factors may affect the immunogenicity of influenza vaccine, including vaccine factors, adjuvants, individual factors, repeated vaccination, and genetic factors.Citation11–14

Effect of vaccine factors

Dosage of vaccines

The effect of vaccine dose on immunogenicity usually varies from population to population. A study showed that antibody responses to half-dose trivalent inactivated vaccine (TIV) in adults were not inferior to the full-dose vaccine, particularly for those aged between 18 and 49. This suggests that under the circumstance of vaccine shortage, half-dose vaccination can be an option for 18–49 adults. However, for people aged 50–64, the antibody responses induced by half-dose vaccine is significantly lower than that by full-dose vaccine, indicating that half-dose vaccine cannot provide enough protection for elderly people.Citation15 A prospective cohort study conducted by Coleman et al.Citation16 also confirmed that, for healthy adults aged 20–45, the immunogenicity was adequate in both half-dose and full-dose AS03-adjuvanted monovalent 2009 pandemic H1N1 vaccine, and the difference has no significance in statistics. Many studies have shown that, compared with standard-dose influenza vaccine, high-dose TIV can effectively improve the antibody responses and reduce laboratory-confirmed influenza-like illness for elderly people.Citation17–19 For children, while exploring the optimal dose of vaccination, the safety of vaccine also should be seriously considered. A study showed that infants aged 6–35 months who received full-dose TIV had higher levels of geometric mean titers against A/H1N1 than those who received half-dose TIV. No differences were found between two dose groups against A/H3N2 and B, and the safety of full-dose TIV was also demonstrated in this study.Citation20 Skowronski et al.Citation21 found that for infants aged 6–11 months, full-dose TIV may induce higher HI antibody titers to all 3 vaccine components compared with half-dose, without increasing reactogenicity. In general, most current studies suggested that high-dose vaccination is correlated with better immunogenicity, many scientists also recommend high-dose vaccines for people with heavy disease burdens, such as the elderly and immunocompromised individuals.Citation22,Citation23 A high-dose influenza vaccine called Fluzone High-Dose Quadrivalent which contains four times the antigen was licensed only for persons aged 65 years and older. This kind of higher-dose vaccine is well tolerated and highly immunogenic, and may provide a better protection for elderly people.

Delivery modes of vaccination

Influenza vaccines can be delivered in a variety of ways, primarily by traditional intramuscular (IM) injection. In 2011–2012, an intradermal (ID) influenza vaccine was first used on people aged 18–64 in the US. A randomized-controlled trial (RCT) showed that the GMT and seroprotection rate of individuals injected ID vaccine were higher than those injected with traditional IM vaccine.Citation24 Another two RCT researches confirmed that the immunogenicity of ID influenza vaccine with 9 μg hemagglutinin (HA) was comparable to IM vaccine with 15 μg HA, and the safety was acceptable.Citation25,Citation26 Compared with traditional IM vaccine, ID vaccine has many advantages including: lower antigen dosage and vaccine cost; application of microinjection system, smaller needle, less pain and simpler control.Citation27 Based on the advantages mentioned above, many experts believe that the use of ID method will help to improve the coverage rate of influenza vaccine. Besides IM and ID injection, influenza vaccine also can be delivered by inhalation. This mode of inoculation could simulate the natural infection process and induce normal immune response, at the same time promote the secretion of immunoglobulin antibodies by the upper respiratory mucosal epithelium. In this way, it will provide a better and wider protection for the inoculators.Citation28 To increase the coverage of influenza vaccination and improve the VE, we still have to explore and develop more delivery methods.

Vaccine types

Currently, there are mainly three types of vaccines in the global market: inactivated influenza vaccine (IIV), live-attenuated influenza vaccine (LAIV), and recombinant influenza vaccine (RIV). Among them, IIVs are the most efficient, valuable, and low‐cost tools to effectively reduce influenza virus infections.

LAIV is a kind of nasal spray vaccine in which the virulence has been weakened but retains immunogenicity. After the inhalation with LAIV, pathogens can grow in the human body and activate the corresponding immune responses without causing any disease. This natural infection pathway can not only induce humoral immunity and cellular immunity, but also induce mucosal immunity to provide a long-term and more extensive immune protection to the body. Compared with IIV, LAIV can also extend the duration of the antibody-secreting cell (ASC) response through the long-term stimulation caused by virus replication.Citation29 However, LAIV is not recommended for children under two years of age, adults aged ≥50, pregnant women, and people with immunodeficiency due to its residual virulence. Nowadays, in terms of the comparison of VE, most studies showed that the VE of LAIV was comparable to TIV and sometimes TIV presented a higher VE in adults, while in children and adolescents, LAIV has a higher VE than TIV.Citation30,Citation31 While in terms of the immunogenicity, most studies suggest that IIV induces a stronger antibody response. An observed study conducted in 2013–2014 influenza season showed that among children aged 5–17, the HI antibody responses of LAIV recipients were lower than that of IIV recipients.Citation32 American researchers found that healthy children aged 3–17 who received IIV had higher postvaccination GMTs and seroconversion rates to all four vaccine components than LAIV recipients.Citation33 Another study of children with cancer confirmed that, compared with LAIV, TIV induced higher GMT against influenza A viruses (P < .001), greater seroprotection against influenza A/H1N1 (P = .01), and greater seroconversion against A/H3N2 (P = .004).Citation34 Similarly, Canadian scientists also concluded that the magnitude of seroconversion induced by IIV against influenza A virus was much greater, suggesting that LAIV was less immunogenic than IIV.Citation35 It is worth mentioning that the immunogenicity of LAIV may be underestimated by using HI titer as an evaluation indicator since the local mucosal immune responses and T cell responses elicited by LAIV cannot be measured by HI titer.Citation29,Citation36 For adults, another possible reason why the immunogenicity of LAIV is underestimated is that the virus can be partly neutralized or eliminated by their previously acquired local secretory IgA (sIgA), serum IgA, serum IgG, and cytotoxic T lymphocytes. sIgA is the main humoral mediator of mucosal immunity, its antibodies can neutralize potential pathogens at the nasal entrance site before they attack the epithelial cells. While serum IgA and IgG antibodies transude from the serum to the mucus and provide protection against infection after the virus invades the mucosal epithelium.Citation37,Citation38 After the infection, the protection mediated by IgA local immune response in the upper respiratory system may last for 3–6 months or longer.Citation29,Citation37,Citation39 As a result, LAIV inoculated during this period will be neutralized to a certain extent before eliciting adaptive immune responses. For most adults who have been infected with influenza, LAIV may also be partly neutralized by tissue-resident memory T cells in lungs and immunological cells which are stored in the tonsil.Citation40,Citation41 Therefore, the effect of LAIV in adults cannot be objectively evaluated by serological indicators alone. Nowadays, since studies on VE and immunogenicity of LAIV are contradictory, the application of LAIV is still restrained and closely monitored in several countries. However, for young children, the priming of T cell responses and mucosal immune responses are particularly important since young children have not been through many infections so they have not acquired many specific memory B cells yet. Mucosal antibodies are essential in protecting the upper respiratory tract from infection. While T cell responses have a variety of functions, including preventing virus replication, participating in antigen presentation, inducing the differentiation of B cells and mediating the humoral immune response. The ability of LAIV to induce a robust T cell responses and mucosal IgA antibodies have been demonstrated.Citation42,Citation43 Hoft et.alCitation44 reported that LAIV and IIV induce similar humoral responses, but only LAIV induces diverse T-cell responses in young children. Therefore, the protective potential of LAIV for children in influenza season is of importance and require further investigation.

RIV is a synthetic vaccine that reassembles the genes of influenza viruses onto some other viruses, plasmids, bacteria,, and cells. Eggs are not required in the whole producing process of RIV. In this way, the manufacturing process might be faster than that of egg-based vaccines. In 2013, a trivalent RIV called FluBlok made from Protein Sciences was first licensed by the FDA. Afterward in 2017, a quadrivalent version was licensed to use on adults 18 years and older in the United States. RIV contains purified recombinant HA protein antigens which use the baculovirus as an expression vector. Its immunogenicity and safety have also been demonstrated in recent years. The HA dose in FluBlok was three times higher than that in IIV without a significant increase in side-effects.Citation45 An earlier phase 1 clinical trial found that the HI titer of trivalent RIV (RIV3) was lower than that of TIV in children aged 6 to 59 months. The serologic responses to FluBlok were higher in the older group (children aged 36–59 months) than the younger group, but were still somewhat lower compared to TIV.Citation46 A randomized trial conducted in subjects aged 6 to 17 years showed that the GMT and seroconversion rate of RIV to H1N1 and H3N2 were higher than that of IIV. No vaccine-related adverse events were found in both two vaccines.Citation47 Baxter et al.Citation48 observed higher antibody responses to influenza A virus, but similar responses to influenza B virus in FluBlok group in comparison to IIV group in healthy adults. The superior immunogenicity of RIV (especially to A/H3N2) has also been reported in the elderly≥65 years.Citation49 In a word, RIV was safe, immunogenic, and even effective under the condition of antigenic mismatch between the vaccine antigens and circulating viruses, suggesting the potential of RIV to provide a better cross-protection.Citation50 With the continuous innovation of technology on vaccines, more and more emerging RIVs such as NanoFlu from Novavax and Supemtek from Sanofi will be available soon.

Effects of adjuvants

The addition of adjuvant is a practical means of increasing the immunogenicity and effectiveness of vaccines. Aluminum adjuvant is one of the most classical influenza vaccine adjuvants. As a kind of adsorbent, aluminum adjuvant may strongly absorb antigens into precipitation and slowly release it to prolong the reaction time and fully activate the humoral immune response, while its ability to induce cellular immune response is poor.Citation51 In order to optimize the immunogenicity of influenza vaccines, various new adjuvants such as oil-in-water emulsions including AS03 and MF59 have emerged. Their role is to induce immune cells to secrete cytokines and chemokines, promote transition from monocytes to dendritic cells and recruit monocytes and neutrophils from the blood to migrate to the vaccination site. Thus, the immune response can be enhanced.Citation52–54 An RCT study designed to evaluate the immunogenicity of an inactivated 2009 H1N1 influenza vaccine showed that in both healthy adults and elderly people, the GMT of subjects in the AS03-adjuvanted group was statistically higher than that in the control group. 61% of adjuvant free subjects and 81% of AS03 adjuvanted subjects reached seroprotective levels, respectively.Citation55 A study of children aged 30 to 96 months showed that compared with recipients who only received TIV, MF59-adjuvanted TIV vaccination produced 6.9 to 8.0-fold higher antibody responses against A/H3N2 and B strains.Citation56 Another study aimed to evaluate the immune responses to A/H1N1 vaccination also reported that the application of MF59 as an adjuvant may significantly upregulate the speed and level of antibody response.Citation57 The superior immunogenicity of MF59-adjuvanted influenza vaccines was also proved by a systematic review and meta-analysis contained 31 studies.Citation58 Many researches have confirmed that influenza vaccines adjuvanted with AS03 or MF59 are generally well tolerated and displayed an acceptable safety profile similar to the nonadjuvanted ones.Citation53,Citation59–61 A MF59 added influenza vaccine named FLUAD was already approved in the United States for persons aged 65 years and older in 2015. Now, its quadrivalent version was also approved in 2020. Studies of some other emerging adjuvants were also performed. As a potential adjuvant for influenza vaccines, CpG can activate immune cells such as T cells, B cells, and NK cells to produce larger amounts of cytokines to enhance the body’s specific and nonspecific immune response. Researchers found that the use of CpG7909 adjuvant may maintain its immunogenicity with lower dose to save the vaccine cost. The CpG7909 adjuvanted vaccine was also found to have similar frequency and intensity of most adverse reactions to nonadjuvanted ones, and both of them were well tolerated.Citation62 The mechanisms of cytokine adjuvants such as interferon depend on their original regulatory functions in the immune system. In principal, adjuvanted influenza vaccines are safe and well-tolerated, though they may increase some local site symptoms like redness and pain at the injection site. Except for FLUAD, other licensed influenza vaccines with adjuvants like Prepandrix, Orniflu, Panflu and so on are still in use under different conditions.Citation63 To cope with different influenza seasons, studies on practical-adjuvanted influenza vaccines still need to be further explored.

Effect of individual factors

Gender

Gender is one of the important factors which may influence the immune responses of influenza vaccine. In Engler’s study,Citation15 women of all ages had been identified with significant higher GMT than man after vaccination, whatever the dose or strain is. Other studies have found that vaccines can induce higher levels of CD4 + lymphocytes and Th1 cytokines in women than men.Citation64,Citation65 In the light of this phenomenon, some researchers believe that the reason why women’s responsiveness is higher than that of men can be attributed to the difference in the sex steroid hormone levels. This kind of hormone can directly binding to intracellular receptors in immune cells, such as monocytes, B cells and T cells.Citation66,Citation67 Furma et al.Citation68 found that men with elevated serum testosterone level and high expression of lipid metabolism-related genes often conduct the lowest antibody responses to TIV, suggesting that testosterone could inhibit the immune response after influenza vaccination. For women, half of the activated genes in T cells have estrogen response elements in their promoters, indicating that estrogens can directly mediate the expression of these genes to influence the activity of T cells.Citation69 In addition, estradiol can also stimulate the production of antibodies by B cells at physiological concentrations.Citation70 American researchers found that after the vaccination, compared with adult males, adult females developed higher vaccine-induced immune responses to the influenza vaccine. And this immune response is positively associated with the concentrations of estradiol. The mice model also showed that the antibody responses were increased in females by estradiol and decreased in males by testosterone.Citation71 In female mice, the ovariectomy may negatively modulate the antibody response to influenza vaccine, but the immune response can be restored after the administration of estrogen.Citation72

Age

Individuals of different ages also differ in their ability to respond to influenza vaccines. Young children, especially newborns, usually have a higher burden of influenza infection and a relatively weak responsiveness to vaccines. It has been reported that children aged 2 to 5 years have a higher immune response to TIV than children under 2 years of age.Citation73 Sasaki et al.Citation74 also found that IgG circulating ASC and serum antibody responses were relatively low in the younger children compared to older children and adults, suggesting that the immune responses induced by vaccines were different in various age groups. The immune system of infants is not fully developed, their magnitude and activity of antigen-presenting cells (APCs), immune cells and cytokines are significantly lower than adults and older children.Citation75 During infancy, the peripheral B cell pool is characterized predominantly by transitional and naive B cells.Citation76 Due to the lack of memory B cells generated from antigenic stimulation, infants are less responsive to vaccines compared with adults and older children. For newborns, the neonatal immunological milieu is polarized toward Th2-type immunity with dampening of Th1-type immunity. Compared with older children, infants’ functional deficiencies of APCs in innate immune system and impaired humoral immunity may result in poorer antibody responses, both in quantity and quality.Citation77,Citation78 Besides, researchers concluded that the low abundance of intestinal microbiota may also account for the relatively weak immunity of infants.Citation79 Old people are also part of the vulnerable populations. Current evidence suggests that influenza vaccines are less effective in older people than in young adults, and the protection period induced by influenza vaccines in the elderly is also relatively short.Citation11,Citation80,Citation81 A retrospective analysis of 31 studies by Goodwin et al.Citation81 showed that the estimate of clinical vaccine efficacy in the elderly (aged 58–104) was significantly lower than that of in young adults (17–53% vs. 70–90%), as well as the seroconversion rate, seroprotection rate and the GMT level. In addition, the serological responses after the vaccination of the elderly over 75 years old is even worse than those under 75 years old. According to the data of 2015–2016 influenza epidemic season in Poland, a study reported that the GMT level and seroprotection rate of children aged 0–4 and the elderly over 65 were both lower, suggesting the susceptibility of these two age groups to influenza.Citation82 In a retrospective study in Scottish, based on data from nine influenza seasons, the VE was 57.1% [95% confidence interval (CI) = 31.3–73.3%] for all age groups and 59.6% (95% CI = 21.9–79.1%) for people under 65 years old. For old people over the age of 65, the VE was only 18.8% (95% CI = −103.7–67.6%), which was much lower than that in the non-elderly groups.Citation83 Unlike young children, the underlying cause of the low responsiveness to vaccines in old people is age-dependent decrease in immunological competence, often referred to as ‘‘immunosenescence’’.Citation84 Aging has a series of negative effect on a variety of molecules, cells, and structures in the body. With increased age, there may be a degeneration of thymus, decrease in phagocytosis, downregulation of antigen presentation, deficiency of B cells and T cells, imbalance of Th1/Th2 cytokines, and progressive deterioration of innate and adaptive immune responses.Citation85–87 Panda et al.Citation88 found that the production of immune-related factors such as TNF-α, IL-6, IL-12, and IFN-α in the elderly (>65 years old) was significantly lower than that in the young adults (21–30 years old), suggesting that aging is associated with the imbalance of cytokine levels and may reduce the immune responses. The effects of immunosenescence have also been demonstrated by numerous studies in mouse model.Citation89,Citation90 In a word, it is of great significance for the public health to popularize the coverage of influenza vaccination among the elderly and provide them with a more optimized vaccination strategy due to their reduced capacity for antiviral infection and poorer responsiveness to vaccines.

Body mass index (BMI)

Sagawa et al.Citation91 found that for people aged over 65 with a BMI<18.5 and a weight loss ≥5% within 6 months had a poor immune response to influenza vaccine. Another study of 48 diabetic patients showed a dose–response relationship between BMI index and serological response to vaccination. The proportion of subjects whose specific antibody level to A/H1N1 after vaccination increased ≥4 times in patients with BMI<22.1, BMI = 22.1–23.8 and BMI>23.9 were 62, 69, and 100%, respectively, and the difference was still statistically significant (P = .008) after adjusting for confounding factors.Citation80 Except for low BMI, high BMI and obesity are also risk factors for low responsiveness to influenza vaccination. It has been reported that compared with individuals with normal body weight, obese individuals (BMI>30) have decreased CD8 + T cell activation, decreased expression of functional proteins, and a steeper decline in vaccine antibody over time, indicating that obesity may impair the body’s ability to induce protective antibodies.Citation92 American researcher HonceCitation93 also found that obesity may not only lead to weakened immune responses, but also is associated with increased risk of influenza infection and serious complications. This perspective has also been validated in mouse models of some related studies.Citation94,Citation95 Based on the study of peripheral blood mononuclear cells cultured in vitro, researchers concluded that obesity is related to the deficiencies in activation and function of CD4 + T cells and CD8 + T cells. Compared with healthy weight individuals, obese people had lower levels of activation markers such as CD69, CD28, CD40 ligand, and IL-12 receptor expressed by CD4 + T cells and CD8 + T cells, and lower levels of functional biomarkers including IFN-γ and granzyme B.Citation96 In addition, differences in the richness and diversity of gut microbiome between obese and healthy people may also contribute to their different responses to vaccines.Citation97

Unhealthy condition

Health status is also a factor that affects the body’s immunity. The comparison of immune response to influenza vaccines in people with different health conditions are listed in . A study of 70 patients with hematological malignancies found that only 39.3% of patients with B-cell malignancies (P < .001), 45.5% with allogeneic stem cell transplant recipients (P < .001) and 85.0% of patients with chronic myeloid leukemia (P = .086) were found to produce protective antibodies, compared with healthy controls whose antibody titers were all reached protective level after vaccination.Citation98 A prospective study on immunocompromised children with autoimmune diseases, AIDS, congenital immunodeficiency, or prematurity from Meier et al.Citation99 showed that the GMT and seroprotection rate of immunocompromised children were lower than that of immunocompetent children. For most organ transplant recipients, they can only induce moderate immune responses after the vaccination. According to a study conducted in 66 stable renal transplant recipients and 19 healthy volunteers, researchers found that compared with healthy volunteers, postvaccinal humoral responses to A/H1N1 and A/H3N2 strains were less frequent in transplanted patients (H1N1: 9.5% vs. 57.9%, P < .001; H3N2: 21.5% vs. 57.9%, P = .004)Citation100; Another study conducted in Germany reported that the influenza vaccine does not provide a protective immune response in the majority of kidney transplant recipientsCitation101; Duchini et al.Citation102 also found that liver transplant recipients were less responsive to A/H3N2 influenza vaccine than healthy adults. For diabetics, some studies have shown that their immune responses to influenza vaccine are comparable to those of normal people.Citation103,Citation104 While analyzing the postvaccinal antibody level and seroprotection rate of 44 patients with Duchenne muscular dystrophy (DMD) and 41 healthy healthcare workers, Japanese researchers concluded that there is no significant difference between these two groups.Citation105 For AIDS patients, study showed that influenza vaccine may induce them to produce antibodies comparable to those of healthy people, suggesting the applicability of influenza vaccine for AIDS patients.Citation106

Table 1. Immune response to influenza vaccines in people with different health conditions

Habits and customs

Living habits such as smoking, drinking, staying up late, diet, and so on have a great impact on the immune system. Godoy et al.Citation107 found that among the elderly over the age of 65, smoking may greatly reduce the VE and was associated with the increased rates of influenza-related hospitalizations (OR = 1.32, 95%CI = 1.04–1.68). The VE was 21% in current smokers/ex-smokers and 39% in nonsmokers. In Spain, a multicenter case–control study conducted in old people also came to a similar conclusion, researchers in this study suggested that those with a history of smoking should be recommended as one of the target populations for routine influenza vaccination. Among adults over 18 years of age, current smokers had a higher risk of influenza-related hospitalization than ex-smokers (OR = 2.18 vs. 1.73), indicating that the health effects of smoking do not disappear, but will decrease over time.Citation108 Horvath et al.Citation109 found that the activity of NK cells and the levels of interferon-γ in smokers were inhibited after influenza vaccination. Experimental animal model studies have also shown that smoking impairs the respiratory immune system, leads to a decrease in cytokines such as interferon and CD4 + T cells, dampens antiviral signaling in small airway epithelial cells, and thus downregulates the body’s ability to defend against pathogens.Citation110–113 Besides smoking, long term and high alcohol consumption is also a risk factor for suppressing the immunity and increasing the body’s susceptibility to pathogens,Citation114 while moderate alcohol consumption was associated with reduced inflammation and better immune responses to some vaccines.Citation115 Researchers found that alcohol may influence the immune system by regulating the balance and interaction of host microbiome.Citation116 At present, although the evidence on the association of lifestyle habits with the immune responses to influenza vaccination is not sufficient, existing studies still suggests that it is necessary to take the habits and customs into consideration while analyzing the effectiveness and immunogenicity of influenza vaccines.

Microbiome

More and more evidence has proven the important role of microbiome in modulating human immunity.Citation117,Citation118 Zimmermann et al.Citation119 found that the adaptive immune responses were positively associated with the relative abundance of the phylum actinobacteria and firmicutes but negatively correlated with the phylum Proteobacteria and Bacteroidetes. As for the antibody responses to influenza vaccines, Thomas Hagan et al.Citation97 found that compared with the control group, the antibiotic group had lower levels of neutralizing antibodies to A/H1N1, indicating that the application of antibiotics may impair the immunity by reducing the richness and diversity of gut microbiome. Another study showed that the serological responses to TIV of germ-free or antibiotic-treated mice were impaired, but can be restored by taking a strain of E. coli orally.Citation120 Toll-like receptor 5 (TLR5) is an important immune-related protein molecule, which is expressed in bone marrow-derived cells such as mononuclear macrophages. Many studies have confirmed the positive correlation between the expression of TLR5 and the antibody responses.Citation120–123 Jason et al.Citation120 found that compared with wild-type mice, TIV-specific IgG, and IgM antibody responses were significantly reduced in Tlr5−/- mice. Bacterial flagellin is the only ligand of TLR5, thus, the microbiome may influence the immune responses to influenza vaccine by regulating the expression of TLR5. Emerging evidence proved that the immunogenicity of influenza vaccines can be improved by using flagellin as an adjuvant.Citation124–128 In addition to the TLR5-mediated pathway, the microbiome may also directly or indirectly affect the antibody responses through a variety of ways, such as hormones and immune-related metabolites.Citation129 To further explore the specific mechanisms of microbiome on immunity, other related factors should also be considered.

Effect of repeated vaccination

For people with low immunity, repeated vaccination in the same influenza season may improve the protective effect of the influenza vaccine to some extent. In the study of 70 patients with hematological malignancies mentioned above, researchers also found a significant increase in the seroprotection rates from 39% to 68% (P = .008) in patients with B-cell malignancies and from 45% to 73% (P = .031) in allogeneic SCT recipients after the second vaccination, suggesting that the booster immunization could significantly improve the antibody level of patients.Citation98 Another study also showed that the second vaccination may upregulate the immunogenicity of influenza vaccine in the elderly aged over 61 years old. The seroprotection rate of subjects was 79.1% 21 days after the first dose, but rose to 93.3% 14 days after the second dose (35 days after the first dose).Citation130 However, unlike repeated vaccination during the same flu season, continuous annual vaccination may lead to reduced VE and immunogenicity of vaccines. When encountering a virus strain similar but not identical to the one previously infected, the immune system tends to induce a strong anamnestic response only to the original-infected strain, and may also diminish the production of memory cells stimulated by the subsequent one.Citation131 Studies reported that prior year vaccination may reduce the effector B-cell responses to new TIV immunization.Citation132,Citation133 In an influenza vaccination cohort study of pregnant women, researchers found that higher baseline antibody levels (P < .001) and prior year influenza vaccination (P = .03) were both statistically associated with reduced odds of seroconversion.Citation134 Thompson et al.Citation135 also concluded that repeated annual vaccination was related to the decrease of geometric mean fold change ratios (GMRs) to A/Perth/16/2009-like virus, suggested that previous vaccination history may have a negative impact on the immune responses of the follow-up vaccination. Surender Khurana et al.Citation136 also confirmed that, regardless of the vaccine platform, the antibody-affinity maturation to HA of all three influenza virus strains can be reduced by repeated annual influenza vaccination. The specific mechanism of how preexisting immunity affects immunogenicity is still unclear and requires further exploration.

Effect of genetic factors

For individuals with basically the same individual factors such as age and sex, even if they are vaccinated with the same vaccine, the immune responses generated following the vaccination are often different, indicating that genetic factors play an important role in this process. Single nucleotide polymorphisms (SNPs), as one of the most common types of heritable variation in humans, has been shown to have a significant impact on the immune responses to vaccines.Citation137–139

SNPs in HLA

HLA is a polygenic and polymorphic complex located on the short arm of chromosome 6, which plays an important role in antigen recognition. After the invasion of pathogens, it is first absorbed and degraded into polypeptides by APC, and then expressed on the APC surface in the form of peptide-HLA (p-HLA) complex, which transmits the information of antigen to CD4 + T cells to initiate the whole immune response process.Citation140,Citation141 A number of studies have reported the relationship between SNPs in HLA and the immune responses to influenza vaccination: As early as 1976, researchers found that the HLA-A Type W16 allele was associated with lower levels of influenza-specific antibodies.Citation142 An RCT study conducted in 185 elderly people from Moss et al.Citation143 found that compared with the elderly who fail to reach the seroprotection level, the frequencies of HLA-DRB1*04:01 allele and HLA-DPB1*04:01 allele were higher in those who reached the seroprotection level. British researchers found an increase in the frequency of HLA-DRB1* 0701 (P = .016) and a decrease in the frequency of HLA-DQB1*0603-9/14 (P = .045) in non-responders who received TIV.Citation14 Based on the analysis of immune responses of healthy Caucasian men aged 18–40 years after influenza vaccination, Poland et al.Citation144 concluded that HLA-A*1101 (P = .0001), HLA-A*6801 (P = .09), HLA-B*3503 (P = .02), HLA-B*1401 (P = .06), HLA-C*0802 (P = .05), HLA-DRB1*1104 (P = .04), HLA-DRB1*1601 (P = .02) and HLA-DQB1*0502 (P = .03) alleles were correlated with a higher median of specific antibodies to H1N1, while the HLA-DRB1* 1303 allele (P = .04) was associated with lower antibody titers to H1N1. It has also been reported that the HLA-DR7/4, DQB1*0302 genotype (OR = 5.15; 95%CI = 1.94–13.67; P = .001) of non-Hispanic white children and the HLA-DR7/Y (including DRB1 11, DRB1 13 and DRB1 14) genotype (OR = 5.84; 95%CI = 1.68–20.28; P = .006) of Hispanic children were correlated with low responsiveness to TIV vaccination.Citation145 Currently, the specific mechanisms by which gene variants in HLA influence the immune responses are not clear. Some scholars speculate that individuals with different HLA genotypes may produce HLA molecules with various antigen affinity through transcription and translation, thereby affecting the efficiency of antigen presentation.

SNPs in cytokines

Cytokines act as cell-signaling molecules between immune cells and play an important role in both innate and adaptive immunity. Human Functional Genomics Project (HFGP) showed that for approximately 70% of peripheral blood mononuclear cells, the effect of genetic factors on their cytokine responses varies from 0.15 to 0.75.Citation146 Many studies have confirmed that SNPs outside HLA region, such as those on cytokines, also have important effects on the immune responses to influenza vaccination.Citation144,Citation147 The production of specific antibodies is a process mediated by lymphocytes and requires the assistance of T helper cells (Th). Cytokines secreted by Th1 cells, such as tumor necrosis factor-α (TNF-α), interleukin-2 (IL-2) and interferon-γ (IFN-γ), are mainly involved in cellular immunity and delayed type hypersensitivity. These cytokines play an important role in the body’s inflammatory responses caused by intracellular microbial infection and cellular immunity. While cytokines secreted by Th2 cells, such as IL-4, IL-5 and IL-10, are mainly involved in humoral immunity and regulate the immune responses against extracellular microbial and parasitic infections, as well as allergic reactions. Poland et al.Citation144 found that IL-6 rs1800796 GG genotype and IL-6 rs2069861 AA genotype were associated with higher antibody titers, while IL-12B rs3212227 CC genotype, TNF receptor superfamily member 1A (TNFRSF1A) rs4149621 G allele, and IL-1 receptor-1 (IL1R1) rs3732131 GG genotype were correlated with lower antibody levels. A study performed from Canadian researchers reported that IL-28B rs8099917 TG+GG genotype was associated with the increase of seroconversion rate after influenza vaccination (OR = 1.99, P = .038), and proposed that IL-28B is a key regulatory factor to maintain the balance of Th1/Th2.Citation147

SNPs in immune-related molecules

The immune system is a complicated system, which requires regulations from a series of immune-related molecules. Genetic variants in these molecules may also influence the immune responses. Leptin is a kind of proteohormone secreted by adipose cells, which can regulate the activity of energy metabolism, reproductive development and immune system. A study from the Mayo Clinic concluded 8 SNPs in leptin and its receptor were statistically correlated with the specific antibodies induced by influenza vaccination.Citation148 Since there is a strong association between the leptin levels and BMI, this study also suggests that obesity may also affect the immunity. Another study on interferon-induced transmembrane protein-3 (IFITM3) showed that the seroconversion rate of subjects with IFITM3 rs12252 CC genotype on day 14 after influenza vaccination was 48.6%(17/35), which was much lower than that of subjects with TT genotype (78.6%, 22/28, P = .015), indicating that TT genotype may provide a better immune protection for its carriers.Citation149 Another study conducted in 171 healthy volunteers with 3 different IFITM3 rs12252 genotypes from Na Lei et al.Citation150 showed that, the seroconversion rate to H1N1, H3N2, and B of IFITM3 rs12252 CC carriers were all lower than that of CT and TT carriers. Subsequently, they also confirmed that IFITM3 deletion attenuated the antibody response to influenza vaccination in mouse model. Heme oxygenase (HO) is an enzyme involves in the physiological activity of various cytokines and immune cells. A cohort of 147 healthy human subjects carried from Cummins et al.Citation151 showed that HMOX1 rs743811 G allele (P = .017) and HMOX2 rs2160567 G allele (P = .014) were both related to poorer vaccine immune response.

The expression and function of HLA, cytokines and other immune-related molecules are all regulated by their gene variants. Nowadays, although a lot of SNPs have been confirmed to affect the immune responses to influenza vaccination, few studies have been conducted at the molecular level. Besides, further genome-wide studies should also be carried out identify more immune-related variants.

Conclusions

Vaccination is the most effective way to prevent and control the prevalence of influenza. However, due to the limited research progress of broad-spectrum universal influenza vaccine and relatively few cases of severe infection, influenza vaccination still has a narrow coverage in the population. Many “vulnerable groups” are still facing a high risk of infection in influenza seasons. In order to provide better protection for people and provide more clues for future research, it is of great significance to fully analyze the relevant factors that may affect the specific antibody responses, to identify low and non-responders to influenza vaccination, to actively develop more efficient new vaccines, and to improve the individualized vaccination program.

Competing interests

None declared.

Additional information

Funding

This work was supported by the Special Funds of the National Natural Science Foundation of China [Grant No. 82041043].

References

  • Hswen Y, Brownstein JS, Liu J, Hawkins JB. Use of a Digital Health Application for Influenza Surveillance in China. Am J Public Health. 2017 Jul;107(7):1130–36. PMID:28520492. doi:10.2105/ajph.2017.303767.
  • Omer SB, Goodman D, Steinhoff MC, Rochat R, Klugman KP, Stoll BJ, Ramakrishnan U. Maternal influenza immunization and reduced likelihood of prematurity and small for gestational age births: a retrospective cohort study. PLoS Med. 2011 May;8(5):e1000441. PMID:21655318. doi:10.1371/journal.pmed.1000441.
  • Mertz D, Geraci J, Winkup J, Gessner BD, Ortiz JR, Loeb M. Pregnancy as a risk factor for severe outcomes from influenza virus infection: A systematic review and meta-analysis of observational studies. Vaccine. 2017 Jan 23;35(4):521–28. doi:10.1016/j.vaccine.2016.12.012. PMID:28024955.
  • Wang GF, Li W, Li K. Acute encephalopathy and encephalitis caused by influenza virus infection. Curr Opin Neurol. 2010 Jun;23(3):305–11. doi:10.1097/WCO.0b013e328338f6c9. PMID:20455276.
  • Togashi T, Matsuzono Y, Narita M, Morishima T. Influenza-associated acute encephalopathy in Japanese children in 1994-2002. Virus Res. 2004 Jul;103(1–2):75–78. PMID:15163492. doi:10.1016/j.virusres.2004.02.016.
  • Britton PN, Dale RC, Blyth CC, Macartney K, Crawford NW, Marshall H, Clark JE, Elliott EJ, Webster RI, Cheng AC, et al. Influenza-associated Encephalitis/Encephalopathy Identified by the Australian Childhood Encephalitis Study 2013-2015. Pediatr Infect Dis J. 2017 Nov;36(11):1021–26. doi:10.1097/inf.0000000000001650. PMID:28654561.
  • Feng L, Shay DK, Jiang Y, Zhou H, Chen X, Zheng Y, Jiang L, Zhang Q, Lin H, Wang S, et al. Influenza-associated mortality in temperate and subtropical Chinese cities, 2003-2008. Bull World Health Organ. 2012 Apr 1;90(4):279–288b. doi:10.2471/blt.11.096958. PMID:22511824.
  • Simonsen L, Viboud C, Taylor RJ. Effectiveness of influenza vaccination. N Engl J Med. 2007 Dec 27;357(26):2729–30. author reply 2730-2721. PMID:18163274..
  • Herrera GA, Iwane MK, Cortese M, Brown C, Gershman K, Shupe A, Averhoff F, Chaves SS, Gargiullo P, Bridges CB. Influenza vaccine effectiveness among 50-64-year-old persons during a season of poor antigenic match between vaccine and circulating influenza virus strains: colorado, United States, 2003-2004. Vaccine . 2007 Jan 2;25(1):154–60. doi:10.1016/j.vaccine.2006.05.129.  PMID:17064823.
  • Morimoto N, Takeishi K. Change in the efficacy of influenza vaccination after repeated inoculation under antigenic mismatch: A systematic review and meta-analysis. Vaccine. 2018 Feb 8;36(7):949–57. doi:10.1016/j.vaccine.2018.01.023. PMID:29373191.
  • Osterholm MT, Kelley NS, Sommer A, Belongia EA. Efficacy and effectiveness of influenza vaccines: a systematic review and meta-analysis. Lancet Infect Dis. 2012 Jan;12(1):36–44. PMID:22032844. doi:10.1016/s1473-3099(11)70295-x.
  • Dominguez A, Godoy P, Torner N. The Effectiveness of Influenza Vaccination in Different Groups. Expert Rev Vaccines. 2016 Jun;15(6):751–64. PMID:26775669. doi:10.1586/14760584.2016.1142878.
  • Castrucci MR. Factors affecting immune responses to the influenza vaccine. Hum Vaccin Immunother. 2018 Mar 4;14(3):637–46. doi:10.1080/21645515.2017.1338547. PMID:28617077.
  • Gelder CM, Lambkin R, Hart KW, Fleming D, Williams OM, Bunce M, Welsh KI, Marshall SE, Oxford J. Associations between human leukocyte antigens and nonresponsiveness to influenza vaccine. J Infect Dis. 2002 Jan 1;185(1):114–17. doi:10.1086/338014. PMID:11756990.
  • Engler RJ, Nelson MR, Klote MM, VanRaden MJ, Huang CY, Cox NJ, Klimov A, Keitel WA, Nichol KL, Carr WW, et al. Half- vs full-dose trivalent inactivated influenza vaccine (2004-2005): age, dose, and sex effects on immune responses. Arch Intern Med. 2008 Dec 8;168(22):2405–14. doi:10.1001/archinternmed.2008.513. PMID:19064822..
  • Coleman BL, Kuster SP, Gubbay J, Scheifele D, Li Y, Low D, Crowcroft N, Mazzulli T, Shi L, Halperin SA, et al. Immunogenicity of a half-dose of adjuvanted 2009 pandemic H1N1 influenza vaccine in adults: a prospective cohort study. European Journal of Clinical Microbiology & Infectious Diseases: Official Publication of the European Society of Clinical Microbiology. 2012 Apr;31(4):591–97. doi:10.1007/s10096-011-1352-5. PMID:21796343.
  • DiazGranados CA, Dunning AJ, Kimmel M, Kirby D, Treanor J, Collins A, Pollak R, Christoff J, Earl J, Landolfi V, et al. Efficacy of high-dose versus standard-dose influenza vaccine in older adults. N Engl J Med. 2014 Aug 14;371(7):635–45. doi:10.1056/NEJMoa1315727. PMID:25119609.
  • Wilkinson K, Wei Y, Szwajcer A, Rabbani R, Zarychanski R, Abou-Setta AM, Mahmud SM. Efficacy and safety of high-dose influenza vaccine in elderly adults: A systematic review and meta-analysis. Vaccine. 2017 May 15;35(21):2775–80. doi:10.1016/j.vaccine.2017.03.092. PMID:28431815.
  • Robison SG, Thomas AR. Assessing the effectiveness of high-dose influenza vaccine in preventing hospitalization among seniors, and observations on the limitations of effectiveness study design. Vaccine. 2018 Oct 29;36(45):6683–87. doi:10.1016/j.vaccine.2018.09.050. PMID:30287157.
  • Halasa NB, Gerber MA, Berry AA, Anderson EL, Winokur P, Keyserling H, Eckard AR, Hill H, Wolff MC, McNeal MM, et al. Safety and Immunogenicity of Full-Dose Trivalent Inactivated Influenza Vaccine (TIV) Compared With Half-Dose TIV Administered to Children 6 Through 35 Months of Age. J Pediatric Infect Dis Soc. 2015 Sep;4(3):214–24. doi:10.1093/jpids/piu061. PMID:26334249.
  • Skowronski DM, Hottes TS, Chong M, De Serres G, Scheifele DW, Ward BJ, Halperin SA, Janjua NZ, Chan T, Sabaiduc S, et al. Randomized controlled trial of dose response to influenza vaccine in children aged 6 to 23 months. Pediatrics. 2011 Aug;128(2):e276–289. doi:10.1542/peds.2010-2777. PMID:21768314.
  • Robertson CA, DiazGranados CA, Decker MD, Chit A, Mercer M, Greenberg DP. Fluzone® High-Dose Influenza Vaccine. Expert Rev Vaccines. 2016 Dec;15(12):1495–505. PMID:27813430. doi:10.1080/14760584.2016.1254044.
  • Hakim H, Allison KJ, Van de Velde LA, Tang L, Sun Y, Flynn PM, McCullers JA. Immunogenicity and safety of high-dose trivalent inactivated influenza vaccine compared to standard-dose vaccine in children and young adults with cancer or HIV infection. Vaccine. 2016 Jun 8;34(27):3141–48. doi:10.1016/j.vaccine.2016.04.053. PMID:27129426.
  • Holland D, Booy R, De Looze F, Eizenberg P, McDonald J, Karrasch J, McKeirnan M, Salem H, Mills G, Reid J, et al. Intradermal influenza vaccine administered using a new microinjection system produces superior immunogenicity in elderly adults: a randomized controlled trial. J Infect Dis. 2008 Sep 1;198(5):650–58. doi:10.1086/590434. PMID:18652550.
  • Gorse GJ, Falsey AR, Fling JA, Poling TL, Strout CB, Tsang PH. Intradermally-administered influenza virus vaccine is safe and immunogenic in healthy adults 18-64 years of age. Vaccine. 2013 May 1;31(19):2358–65. doi:10.1016/j.vaccine.2013.03.008.  PMID:23499604.
  • Gorse GJ, Falsey AR, Johnson CM, Morrison D, Fried DL, Ervin JE, Greenberg DP, Ozol-Godfrey A, Landolfi V, Tsang PH. Safety and immunogenicity of revaccination with reduced dose intradermal and standard dose intramuscular influenza vaccines in adults 18-64 years of age. Vaccine. 2013 Dec 5;31(50):6034–40. doi:10.1016/j.vaccine.2013.09.012.  PMID:24055306.
  • Loubet P, Loulergue P, Galtier F, Launay O. Seasonal influenza vaccination of high-risk adults. Expert Rev Vaccines. 2016 Dec;15(12):1507–18. PMID:27169689. doi:10.1080/14760584.2016.1188696.
  • Sano K, Ainai A, Suzuki T, Hasegawa H. The road to a more effective influenza vaccine: up to date studies and future prospects. Vaccine. 2017 Sep 25;35(40):5388–95. doi:10.1016/j.vaccine.2017.08.034. PMID:28866292.
  • Mohn KG, Smith I, Sjursen H, Cox RJ. Immune responses after live attenuated influenza vaccination. Hum Vaccin Immunother. 2018 Mar 4;14(3):571–78. doi:10.1080/21645515.2017.1377376. PMID:28933664.
  • Heikkinen T, Heinonen S. Effectiveness and safety of influenza vaccination in children. European Perspective Vaccine. 2011 Oct 6;29(43):7529–34. doi:10.1016/j.vaccine.2011.08.011. PMID:21820481.
  • Ambrose CS, Levin MJ, Belshe RB. The relative efficacy of trivalent live attenuated and inactivated influenza vaccines in children and adults. Influenza Other Respi Viruses. 2011 Mar;5(2):67–75. PMID:21306569. doi:10.1111/j.1750-2659.2010.00183.x.
  • King JP, McLean HQ, Meece JK, Levine MZ, Spencer SM, Flannery B, Belongia EA. Vaccine failure and serologic response to live attenuated and inactivated influenza vaccines in children during the 2013-2014 season. Vaccine. 2018 Feb 21;36(9):1214–19. doi:10.1016/j.vaccine.2018.01.016. PMID:29395525.
  • Levine MZ, Martin JM, Gross FL, Jefferson S, Cole KS, Archibald CA, Nowalk MP, Susick M, Moehling K, Spencer S, et al. Neutralizing Antibody Responses to Antigenically Drifted Influenza A(H3N2) Viruses among Children and Adolescents following 2014-2015 Inactivated and Live Attenuated Influenza Vaccination. Clinical and Vaccine Immunology CVI. 2016 Oct;23(10):831–39. doi:10.1128/cvi.00297-16. PMID:27558294.
  • Carr S, Allison KJ, Van De Velde LA, Zhang K, English EY, Iverson A, Daw NC, Howard SC, Navid F, Rodriguez-Galindo C, et al. Safety and immunogenicity of live attenuated and inactivated influenza vaccines in children with cancer. J Infect Dis. 2011 Nov 15;204(10):1475–82. doi:10.1093/infdis/jir561. PMID:21949042.
  • Ang JC, Wang B, Wang JJF, Zeng PYF. Comparative Immunogenicity of the 2014-2015 Northern Hemisphere Trivalent IIV and LAIV against Influenza A Viruses in Children. Vaccines (Basel). 2019 Aug 12;7(3):87. doi:10.3390/vaccines7030087. PMID:31408963.
  • Bandell A, Woo J, Coelingh K. Protective efficacy of live-attenuated influenza vaccine (multivalent, Ann Arbor strain): a literature review addressing interference. Expert Rev Vaccines. 2011 Aug;10(8):1131–41. doi:10.1586/erv.11.73. PMID:21854309.
  • Gianchecchi E, Manenti A, Kistner O, Trombetta C. How to assess the effectiveness of nasal influenza vaccines? Role and measurement of sIgA in mucosal secretions. Influenza Other Respir Viruses. Sep 2019;135:429–37. doi:10.1111/irv.12664. PMID:31225704
  • Leong KW, Ding JL. The unexplored roles of human serum IgA. DNA Cell Biol. 2014 Dec;33(12):823–29. PMID:25188736. doi:10.1089/dna.2014.2639.
  • Brokstad KA, Cox RJ, Eriksson JC, Olofsson J, Jonsson R, Davidsson A. High prevalence of influenza specific antibody secreting cells in nasal mucosa. Scand J Immunol. 2001 Jul-Aug;54(1–2):243–7. doi:10.1046/j.1365-3083.2001.00947.x. PMID:11439173.
  • Zens KD, Chen JK, Farber DL. Vaccine-generated lung tissue-resident memory T cells provide heterosubtypic protection to influenza infection. JCI Insight. 2016 Jul 7;1(10). doi:10.1172/jci.insight.85832. PMID:27468427.
  • Panapasa JA, Cox RJ, Mohn KG, Aqrawi LA, Brokstad KA. The expression of B & T cell activation markers in children’s tonsils following live attenuated influenza vaccine. Hum Vaccin Immunother. 2015;11(7):1663–72. doi:10.1080/21645515.2015.1032486. PMID:26148331.
  • Mohn KG, Brokstad KA, Pathirana RD, Bredholt G, Jul-Larsen Å, Trieu MC, Lartey SL, Montomoli E, Tøndel C, Aarstad HJ, et al. Live Attenuated Influenza Vaccine in Children Induces B-Cell Responses in Tonsils. J Infect Dis. 2016 Sep 1;214(5):722–31. doi:10.1093/infdis/jiw230. PMID:27247344.
  • Mohn KG, Bredholt G, Brokstad KA, Pathirana RD, Aarstad HJ, Tøndel C, Cox RJ. Longevity of B-cell and T-cell responses after live attenuated influenza vaccination in children. J Infect Dis. 2015 May 15;211(10):1541–49. doi:10.1093/infdis/jiu654. PMID:25425696.
  • Hoft DF, Babusis E, Worku S, Spencer CT, Lottenbach K, Truscott SM, Abate G, Sakala IG, Edwards KM, Creech CB, et al. Live and inactivated influenza vaccines induce similar humoral responses, but only live vaccines induce diverse T-cell responses in young children. J Infect Dis. 2011 Sep 15;204(6):845–53. doi:10.1093/infdis/jir436. PMID:21846636..
  • Cox MM, Hollister JR. FluBlok, a next generation influenza vaccine manufactured in insect cells. Biologicals: Journal of the International Association of Biological Standardization. 2009 Jun;37(3):182–89. PMID:19297194. doi:10.1016/j.biologicals.2009.02.014.
  • King JC Jr., Cox MM, Reisinger K, Hedrick J, Graham I, Patriarca P. Evaluation of the safety, reactogenicity and immunogenicity of FluBlok trivalent recombinant baculovirus-expressed hemagglutinin influenza vaccine administered intramuscularly to healthy children aged 6-59 months. Vaccine . 2009 Nov 5;27(47):6589–94. doi:10.1016/j.vaccine.2009.08.032. PMID:19716456.
  • Dunkle LM, Izikson R, Patriarca PA, Goldenthal KL, Cox M, Treanor JJ. Safety and Immunogenicity of a Recombinant Influenza Vaccine: A Randomized Trial Pediatrics. May 2018;141(5). PMID:29610401. doi:10.1542/peds.2017-3021.
  • Baxter R, Patriarca PA, Ensor K, Izikson R, Goldenthal KL, Cox MM. Evaluation of the safety, reactogenicity and immunogenicity of FluBlok® trivalent recombinant baculovirus-expressed hemagglutinin influenza vaccine administered intramuscularly to healthy adults 50-64 years of age. Vaccine. 2011 Mar 9;29(12):2272–78. doi:10.1016/j.vaccine.2011.01.039. PMID:21277410.
  • Treanor JJ, Schiff GM, Couch RB, Cate TR, Brady RC, Hay CM, Wolff M, She D, Cox MM. Dose-related safety and immunogenicity of a trivalent baculovirus-expressed influenza-virus hemagglutinin vaccine in elderly adults. J Infect Dis. 2006 May 1;193(9):1223–28. doi:10.1086/503050. PMID:16586358.
  • Treanor JJ, El Sahly H, King J, Graham I, Izikson R, Kohberger R, Patriarca P, Cox M. Protective efficacy of a trivalent recombinant hemagglutinin protein vaccine (FluBlok®) against influenza in healthy adults: a randomized, placebo-controlled trial. Vaccine. 2011 Oct 13;29(44):7733–39. doi:10.1016/j.vaccine.2011.07.128. PMID:21835220.
  • Xu H, Ruwona TB, Thakkar SG, Chen Y, Zeng M, Cui Z. Nasal aluminum (oxy)hydroxide enables adsorbed antigens to induce specific systemic and mucosal immune responses. Hum Vaccin Immunother. 2017 Nov 2;13(11):2688–94. doi:10.1080/21645515.2017.1365995. PMID:28933668.
  • Domnich A, Arata L, Amicizia D, Puig-Barbera J, Gasparini R, Panatto D. Effectiveness of MF59-adjuvanted seasonal influenza vaccine in the elderly: A systematic review and meta-analysis. Vaccine. PMID:28024956. 2017 Jan 23;35(4):513–20. doi:10.1016/j.vaccine.2016.12.011.
  • Cohet C, van der Most R, Bauchau V, Bekkat-Berkani R, Doherty TM, Schuind A. Tavares Da Silva F, Rappuoli R, Garçon N, Innis BL. Safety of AS03-adjuvanted influenza vaccines: A review of the evidence. Vaccine. 2019 May 21;37(23):3006–21. doi:10.1016/j.vaccine.2019.04.048. PMID:31031030.
  • Wilkins AL, Kazmin D, Napolitani G, Clutterbuck EA, Pulendran B, Siegrist CA, Pollard AJ. AS03- and MF59-Adjuvanted Influenza Vaccines in Children. Front Immunol. 2017;8::1760. doi:10.3389/fimmu.2017.01760. PMID:29326687.
  • Jackson LA, Chen WH, Stapleton JT, Dekker CL, Wald A, Brady RC, Edupuganti S, Winokur P, Mulligan MJ, Keyserling HL, et al. Immunogenicity and safety of varying dosages of a monovalent 2009 H1N1 influenza vaccine given with and without AS03 adjuvant system in healthy adults and older persons. J Infect Dis. 2012 Sep 15;206(6):811–20. doi:10.1093/infdis/jis427. PMID:22782949.
  • Vesikari T, Forsten A, Arora A, Tsai T, Clemens R. Influenza vaccination in children primed with MF59-adjuvanted or non-adjuvanted seasonal influenza vaccine. Hum Vaccin Immunother. 2015;11(8):2102–12. doi:10.1080/21645515.2015.1044167. PMID:26091244.
  • Clark TW, Pareek M, Hoschler K, Dillon H, Nicholson KG, Groth N, Stephenson I. Trial of 2009 influenza A (H1N1) monovalent MF59-adjuvanted vaccine. N Engl J Med. 2009 Dec 17;361(25):2424–35. doi:10.1056/NEJMoa0907650. PMID:19745215.
  • Yang J, Zhang J, Han T, Liu C, Li X, Yan L, Yang B, Yang X. Effectiveness, immunogenicity, and safety of influenza vaccines with MF59 adjuvant in healthy people of different age groups: A systematic review and meta-analysis. Medicine. 2020 Feb;99(7):e19095. PMID:32049815. doi:10.1097/md.0000000000019095.
  • Patel SS, Bizjajeva S, Heijnen E, Oberye J. MF59-adjuvanted seasonal trivalent inactivated influenza vaccine: safety and immunogenicity in young children at risk of influenza complications. International Journal of Infectious Diseases. 2019 Aug;85S:S18–S25. doi:10.1016/j.ijid.2019.04.023. PMID:31051279.
  • Lindert K, Leav B, Heijnen E, Barrett J, Nicolay U, Cumulative clinical experience with MF59-adjuvanted trivalent seasonal influenza vaccine in young children and adults 65 years of age and older. International Journal of Infectious Diseases: IJID: Official Publication of the International Society for Infectious Diseases. Aug 2019;85s;S10–s17. PMID:30904674. doi:10.1016/j.ijid.2019.03.020.
  • Patel SS, Bizjajeva S, Lindert K, Heijnen E, Oberye J. Cumulative clinical experience with MF59-adjuvanted trivalent seasonal influenza vaccine in young children. International Journal of Infectious Diseases. 2019 Aug;85S:S26-S38. doi:10.1016/j.ijid.2019.05.009. PMID:31096055.
  • Cooper CL, Davis HL, Morris ML, Efler SM, Krieg AM, Li Y, Laframboise C, Al Adhami MJ, Khaliq Y, Seguin I, et al. Safety and immunogenicity of CPG 7909 injection as an adjuvant to Fluarix influenza vaccine. Vaccine. 2004 Aug 13;22(23–24):3136–43. doi:10.1016/j.vaccine.2004.01.058. PMID:15297066.
  • Tregoning JS, Russell RF, Kinnear E. Adjuvanted influenza vaccines. Hum Vaccin Immunother. 2018 Mar 4;14(3):550–64. doi:10.1080/21645515.2017.1415684. PMID:29232151.
  • 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 Dec;1(12):1279–83. doi:10.1038/nm1295-1279. PMID:7489409.
  • Huygen K, Palfliet K. Strain variation in interferon gamma production of BCG-sensitized mice challenged with PPD II. Importance of One Major Autosomal Locus and Additional Sexual Influences Cellular Immunology. 1984 Apr 15;85(1):75–81. PMID:6424949.
  • Giefing-Kroll 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 Jun;14(3):309–21. PMID:25720438. doi:10.1111/acel.12326.
  • Klein SL, Flanagan KL. Sex differences in immune responses. Nat Rev Immunol. Oct 2016;1610:626–38. 10.1038/nri.2016.90. PMID:27546235.
  • 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 Jan 14;111(2):869–74. doi:10.1073/pnas.1321060111. PMID:24367114.
  • Hewagama A, Patel D, Yarlagadda S, Strickland FM, Richardson BC. Stronger inflammatory/cytotoxic T-cell response in women identified by microarray analysis. Genes Immun Jul 2009;105:509–16. 10.1038/gene.2009.12. PMID:19279650
  • 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 Apr;128(1):10–20. PMID:11982585. doi:10.1046/j.1365-2249.2002.01780.x.
  • Potluri T, Fink AL, Sylvia KE, Dhakal S. Age-associated changes in the impact of sex steroids on influenza vaccine responses in males and females. NPJ Vaccines. 2019;4:29. PMID:31312529. doi:10.1038/s41541-019-0124-6.
  • 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 Mar 21;29(14):2515–18. doi:10.1016/j.vaccine.2011.01.080. PMID:21310192.
  • WHO. Vaccines against influenza WHO position paper - November 2012. Releve epidemiologique hebdomadaire. 2012 Nov 23;87(47):461–76. English, French.  PMID:23210147.
  • Sasaki S, Jaimes MC, Holmes TH, Dekker CL, Mahmood K, Kemble GW, Arvin AM, Greenberg HB. Comparison of the influenza virus-specific effector and memory B-cell responses to immunization of children and adults with live attenuated or inactivated influenza virus vaccines. J Virol. 2007 Jan;81(1):215–28. PMID:17050593. doi:10.1128/jvi.01957-06.
  • Zhang X, Zhivaki D, Lo-Man R. Unique aspects of the perinatal immune system. Nat Rev Immunol. 2017 Aug;17(8):495–507. PMID:28627520. doi:10.1038/nri.2017.54.
  • Morbach H, Eichhorn EM, Liese JG, Girschick HJ. Reference values for B cell subpopulations from infancy to adulthood. Clin Exp Immunol. 2010 Nov;162(2):271–79. doi:10.1111/j.1365-2249.2010.04206.x. PMID:20854328.
  • Saso A, Kampmann B. Vaccine responses in newborns. Semin Immunopathol. 2017 Nov;39(6):627–42. PMID:29124321. doi:10.1007/s00281-017-0654-9.
  • Yu JC, Khodadadi H, Malik A, Davidson B, Salles ÉDSL, Bhatia J, Hale VL, Baban B. Innate Immunity of Neonates and Infants. Front Immunol. 2018;9:1759. doi:10.3389/fimmu.2018.01759. PMID:30105028.
  • Adlerberth I. Factors influencing the establishment of the intestinal microbiota in infancy. Nestle Nutrition workshop series. Paediatric Programme. 2008;62:13–29;discussion 29–33. doi:10.1159/000146245. PMID:18626190.
  • Egawa Y, Ohfuji S, Fukushima W, Yamazaki Y, Morioka T, Emoto M, Maeda K, Inaba M, Hirota Y. Immunogenicity of influenza A(H1N1)pdm09 vaccine in patients with diabetes mellitus: with special reference to age, body mass index, and HbA1c. Hum Vaccin Immunother. 2014;10(5):1187–94. doi:10.4161/hv.28252. PMID:24717236.
  • Goodwin K, Viboud C, Simonsen L. Antibody response to influenza vaccination in the elderly: a quantitative review. Vaccine. 2006;24(8):1159–69. doi:10.1016/j.vaccine.2005.08.105.
  • Kowalczyk D, Szymański K, Cieślak K, Hallmann-Szelińska E, Brydak LB. Circulation of Influenza Virus in the 2015/2016 Epidemic Season in Poland: serological Evaluation of Anti-hemagglutinin Antibodies. Adv Exp Med Biol . 2019;1150:77–82. doi:10.1007/5584_2018_271. PMID:30276725.
  • Simpson CR, Lone N, Kavanagh K, Ritchie LD, Robertson C, Sheikh A, McMenamin J. Health Services and Delivery Research. Seasonal Influenza Vaccine Effectiveness (SIVE): an observational retrospective cohort study - exploitation of a unique community-based national-linked database to determine the effectiveness of the seasonal trivalent influenza vaccine. Southampton (UK): NIHR Journals Library; 2013.
  • Sadighi Akha AA. Aging and the immune system: an overview. J Immunol Methods. 2018 Dec;463:21–26. doi:10.1016/j.jim.2018.08.005. PMID:30114401.
  • Pawelec G. Age and immunity: what is “immunosenescence”? Exp Gerontol. 2017;105:4–9. doi:10.1016/j.exger.2017.10.024.
  • Crooke SN, Ovsyannikova IG, Poland GA, Kennedy RB. Immunosenescence and human vaccine immune responses. Immunity & Ageing. 2019;16:1–16.
  • Agarwal S, Busse PJ. Innate and adaptive immunosenescence. Annals of allergy, asthma & immunology. Official Publication of the American College of Allergy, Asthma, & Immunology. 2010 Mar;104(3):183–190;quiz 190–182, 210. PMID:20377107. doi:10.1016/j.anai.2009.11.009.
  • Panda A, Qian F, Mohanty S, van Duin D, Newman FK, Zhang L, Chen S, Towle V, Belshe RB, Fikrig E, et al. Age-associated decrease in TLR function in primary human dendritic cells predicts influenza vaccine response. Journal of Immunology (Baltimore, Md : 1950). 2010 Mar 1;184(5):2518–27. doi:10.4049/jimmunol.0901022. PMID:20100933.
  • Goronzy JJ, Weyand CM. Understanding immunosenescence to improve responses to vaccines. Nat Immunol. 2013 May;14(5):428–36. PMID:23598398. doi:10.1038/ni.2588.
  • Haynes L, Lefebvre JS. Age-related Deficiencies in Antigen-Specific CD4 T cell Responses: lessons from Mouse Models. Aging Dis. 2011 Oct;2(5):374–81. PMID:22396889.
  • Sagawa M, Kojimahara N, Otsuka N, Kimura M, Yamaguchi N. Immune response to influenza vaccine in the elderly: association with nutritional and physical status. Geriatr Gerontol Int. 2011 Jan;11(1):63–68. PMID:20738411. doi:10.1111/j.1447-0594.2010.00641.x.
  • Sheridan PA, Paich HA, Handy J, Karlsson EA, Hudgens MG, Sammon AB, Holland LA, Weir S, Noah TL, Beck MA. Obesity is associated with impaired immune response to influenza vaccination in humans. Int J Obes. 2005 [Aug 2012];36(8):1072–77. doi:10.1038/ijo.2011.208. PMID:22024641.
  • Honce R, Schultz-Cherry S. Influenza in obese travellers: increased risk and complications, decreased vaccine effectiveness. J Travel Med. 2019 May 10;26(3). doi:10.1093/jtm/taz020. PMID:30924873.
  • Rebeles J, Green WD, Alwarawrah Y, Nichols AG, Eisner W, Danzaki K, MacIver NJ, Beck MA. Obesity-Induced Changes in T-Cell Metabolism Are Associated With Impaired Memory T-Cell Response to Influenza and Are Not Reversed With Weight Loss. J Infect Dis PMID:30535161. 2019 Apr 19;219(10):1652–61. doi:10.1093/infdis/jiy700.
  • Zhang AJ, To KK, Li C, Lau CC, Poon VK, Chan CC, Zheng BJ, Hung IF, Lam KS, Xu A, et al. Leptin mediates the pathogenesis of severe 2009 pandemic influenza A(H1N1) infection associated with cytokine dysregulation in mice with diet-induced obesity. J Infect Dis. 2013 Apr 15;207(8):1270–80. doi:10.1093/infdis/jit031. PMID:23325916..
  • Paich HA, Sheridan PA, Handy J, Karlsson EA, Schultz-Cherry S, Hudgens MG, Noah TL, Weir SS, Beck MA. Overweight and obese adult humans have a defective cellular immune response to pandemic H1N1 influenza A virus. Obesity (Silver Spring, Md). 2013 Nov;21(11):2377–86 . doi:10.1002/oby.20383. PMID:23512822.
  • Hagan T, Cortese M, Rouphael N, Boudreau C, Linde C, Maddur MS, Das J, Wang H, Guthmiller J, Zheng NY, et al. Antibiotics-Driven Gut Microbiome Perturbation Alters Immunity to Vaccines in Humans. Cell. 2019 Sep 5;178(6):1313–1328.e1313. doi:10.1016/j.cell.2019.08.010. PMID:31491384.
  • de Lavallade H, Garland P, Sekine T, Hoschler K, Marin D, Stringaris K, Loucaides E, Howe K, Szydlo R, Kanfer E, et al. Repeated vaccination is required to optimize seroprotection against H1N1 in the immunocompromised host. Haematologica. 2011 Feb;96(2):307–14. doi:10.3324/haematol.2010.032664. PMID:20971824.
  • Meier S, Bel M, L’Huillier A, Crisinel PA, Combescure C, Kaiser L, Grillet S, Posfay-Barbe K, Siegrist CA. Antibody responses to natural influenza A/H1N1/09 disease or following immunization with adjuvanted vaccines, in immunocompetent and immunocompromised children. Vaccine. 2011 Apr 27;29(19):3548–57. doi:10.1016/j.vaccine.2011.02.094. PMID:21419775.
  • Candon S, Thervet E, Lebon P, Suberbielle C, Zuber J, Lima C, Charron D, Legendre C, Chatenoud L. Humoral and cellular immune responses after influenza vaccination in kidney transplant recipients. Am J Transplant. 2009 Oct;9(10):2346–54. PMID:19656126. doi:10.1111/j.1600-6143.2009.02787.x.
  • Brakemeier S, Schweiger B, Lachmann N, Glander P, Schonemann C, Diekmann F, Neumayer HH, Budde K. Immune response to an adjuvanted influenza A H1N1 vaccine (Pandemrix((R))) in renal transplant recipients. Nephrology, Dialysis, Transplantation: Official Publication of the European Dialysis and Transplant Association - European Renal Association. 2012 Jan;27(1):423–28 . doi:10.1093/ndt/gfr278. PMID:21613386
  • Duchini A, Hendry RM, Nyberg LM, Viernes ME, Pockros PJ. Immune response to influenza vaccine in adult liver transplant recipients. Liver Transplantation: Official Publication of the American Association for the Study of Liver Diseases and the International Liver Transplantation Society. 2001 Apr;7(4):311–13. PMID:11303290. doi:10.1053/jlts.2001.23010.
  • Diepersloot RJ, Bouter KP, Beyer WE, Hoekstra JB, Masurel N. Humoral immune response and delayed type hypersensitivity to influenza vaccine in patients with diabetes mellitus. Diabetologia. 1987 Jun;30(6):397–401. doi:10.1007/BF00292541. PMID:3678660.
  • el-Madhun AS, Cox RJ, Seime A, Sovik O, Haaheim LR. Systemic and local immune responses after parenteral influenza vaccination in juvenile diabetic patients and healthy controls: results from a pilot study. Vaccine. 1998 Jan-Feb;16(2–3):156–60. doi:10.1016/S0264-410X(97)88328-4. PMID:9607024.
  • Saito T, Ohfuji S, Matsumura T, Saito T, Maeda K, Maeda A, Fukushima W, Fujimura H, Sakoda S, Hirota Y. Safety of a Pandemic Influenza Vaccine and the Immune Response in Patients with Duchenne Muscular Dystrophy. Internal Medicine (Tokyo, Japan). 2015;54(10):1199–205. doi:10.2169/internalmedicine.54.1186. PMID:25986256.
  • Agrati C, Gioia C, Castilletti C, Lapa D, Berno G, Puro V, Carletti F, Cimini E, Nisii C, Castellino F, et al. Cellular and humoral immune responses to pandemic influenza vaccine in healthy and in highly active antiretroviral therapy-treated HIV patients. AIDS Res Hum Retroviruses. 2012 Dec;28(12):1606–16. doi:10.1089/aid.2011.0371. PMID:22439734.
  • Godoy P, Castilla J, Soldevila N, Mayoral JM, Toledo D, Martin V, Astray J, Egurrola M, Morales-Suarez-Varela M, Dominguez A. Smoking may increase the risk of influenza hospitalization and reduce influenza vaccine effectiveness in the elderly. Eur J Public Health. 2018 Feb 1;28(1):150–55. doi:10.1093/eurpub/ckx130. PMID:29020390.
  • Godoy P, Castilla J, Mayoral JM, Delgado-Rodriguez M, Martin V, Astray J, Soldevila N, Gonzalez-Candelas F, Castro A, Baricot M, et al. Smoking may increase the risk of hospitalization due to influenza. Eur J Public Health. 2016 Oct;26(5):882–87. doi:10.1093/eurpub/ckw036. PMID:27085194.
  • Horvath KM, Herbst M, Zhou H, Zhang H, Noah TL, Jaspers I. Nasal lavage natural killer cell function is suppressed in smokers after live attenuated influenza virus. Respir Res. 2011 Aug 4;12(1):102. doi:10.1186/1465-9921-12-102. PMID:21816072.
  • Tejero JD, Armand NC, Finn CM, Dhume K, Strutt TM, Chai KX, Chen LM, McKinstry KK. Cigarette smoke extract acts directly on CD4 T cells to enhance Th1 polarization and reduce memory potential. Cell Immunol. 2018 Sep;331:121–29. doi:10.1016/j.cellimm.2018.06.005. PMID:29935764.
  • Bhat TA, Kalathil SG, Bogner PN, Miller A, Lehmann PV, Thatcher TH. Secondhand Smoke Induces Inflammation and Impairs Immunity to Respiratory Infections. J Immunol. 2018 Apr 15;200(8);2927–40. doi:10.4049/jimmunol.1701417. PMID:29555783.
  • Duffney PF, McCarthy CE, Nogales A, Thatcher TH. Cigarette smoke dampens antiviral signaling in small airway epithelial cells by disrupting TLR3 cleavage. Am J Physiol Lung Cell Mol Physiol. 2018 Mar 1;314(3);L505–l513. doi:10.1152/ajplung.00406.2017. PMID:29351447.
  • HuangFu WC, Liu J, Harty RN, Fuchs SY. Cigarette smoking products suppress anti-viral effects of Type I interferon via phosphorylation-dependent downregulation of its receptor. FEBS Lett. 2008 Sep 22;582(21–22):3206–10. doi:10.1016/j.febslet.2008.08.013. PMID:18722370.
  • Szabo G, Saha B. Alcohol’s Effect on Host Defense. Alcohol Research: Current Reviews. 2015;37(2):159–70. PMID:26695755.
  • Barr T, Helms C, Grant K, Messaoudi I. Opposing effects of alcohol on the immune system. Prog Neuropsychopharmacol Biol Psychiatry. 2016 Feb 4;65:242–51. doi:10.1016/j.pnpbp.2015.09.001. PMID:26375241.
  • Chen P, Schnabl B. Host-microbiome interactions in alcoholic liver disease. Gut Liver. 2014 May;8(3):237–41. PMID:24827618. doi:10.5009/gnl.2014.8.3.237.
  • Jamieson AM. Influence of the microbiome on response to vaccination. Hum Vaccin Immunother. 2015;11(9):2329–31. doi:10.1080/21645515.2015.1022699. PMID:26090701.
  • Negi S, Das DK, Pahari S, Nadeem S, Agrewala JN. Potential Role of Gut Microbiota in Induction and Regulation of Innate Immune Memory. Front Immunol. 2019;10:2441. doi:10.3389/fimmu.2019.02441. PMID:31749793.
  • Zimmermann P, Curtis N. The influence of the intestinal microbiome on vaccine responses. Vaccine . 2018 Jul 16;36(30):4433–39. doi:10.1016/j.vaccine.2018.04.066. PMID:29909134.
  • Oh JZ, Ravindran R, Chassaing B, Carvalho FA, Maddur MS, Bower M, Hakimpour P, Gill KP, Nakaya HI, Yarovinsky F, et al. TLR5-mediated sensing of gut microbiota is necessary for antibody responses to seasonal influenza vaccination. Immunity. 2014 Sep 18;41(3):478–92. doi:10.1016/j.immuni.2014.08.009. PMID:25220212.
  • Mukherjee S, Huda S, Sinha Babu SP. Toll-like receptor polymorphism in host immune response to infectious diseases. A Review. 2019 Jul;90(1):e12771. PMID:31054156. doi:10.1111/sji.12771.
  • Didierlaurent A, Ferrero I, Otten LA, Dubois B, Reinhardt M, Carlsen H, Blomhoff R, Akira S, Kraehenbuhl JP, Sirard JC Flagellin promotes myeloid differentiation factor 88-dependent development of Th2-type response. Journal of Immunology. (Baltimore, Md.: 1950).Jun1 2004;17211:6922–30. doi:10.4049/jimmunol.172.11.6922. PMID:15153511.
  • Yang J, Yan H. TLR5: beyond the recognition of flagellin. Cell Mol Immunol. 2017 Dec;14(12):1017–19. PMID:29151579. doi:10.1038/cmi.2017.122.
  • Hong SH, Byun YH, Nguyen CT, Kim SY, Seong BL, Park S, Woo GJ, Yoon Y, Koh JT, Fujihashi K, et al. Intranasal administration of a flagellin-adjuvanted inactivated influenza vaccine enhances mucosal immune responses to protect mice against lethal infection. Vaccine. 2012 Jan 5;30(2):466–74. doi:10.1016/j.vaccine.2011.10.058. PMID:22051136.
  • Kim JR, Holbrook BC, Hayward SL, Blevins LK, Jorgensen MJ, Kock ND, De Paris K, D’Agostino RB Jr., Aycock ST, Mizel SB, et al. Inclusion of Flagellin during Vaccination against Influenza Enhances Recall Responses in Nonhuman Primate Neonates. J Virol. 2015 Jul;89(14):7291–303. doi:10.1128/jvi.00549-15. PMID:25948746.
  • Treanor JJ, Taylor DN, Tussey L, Hay C, Nolan C, Fitzgerald T, Liu G, Kavita U, Song L, Dark I, et al. Safety and immunogenicity of a recombinant hemagglutinin influenza-flagellin fusion vaccine (VAX125) in healthy young adults. Vaccine. 2010 Dec 6;28(52):8268–74. doi:10.1016/j.vaccine.2010.10.009. PMID:20969925.
  • Taylor DN, Treanor JJ, Strout C, Johnson C, Fitzgerald T, Kavita U, Ozer K, Tussey L, Shaw A. Induction of a potent immune response in the elderly using the TLR-5 agonist, flagellin, with a recombinant hemagglutinin influenza-flagellin fusion vaccine (VAX125, STF2.HA1 SI). Vaccine. 2011 Jul 12;29(31):4897–902. doi:10.1016/j.vaccine.2011.05.001. PMID:21596084.
  • Cui B, Liu X, Fang Y, Zhou P, Zhang Y, Wang Y. Flagellin as a vaccine adjuvant. Expert Rev Vaccines. 2018 Apr;17(4):335–49. PMID:29580106. doi:10.1080/14760584.2018.1457443.
  • Ciabattini A, Olivieri R, Lazzeri E, Medaglini D. Role of the Microbiota in the Modulation of Vaccine Immune Responses. Front Microbiol. 2019;10:1305. doi:10.3389/fmicb.2019.01305. PMID:31333592.
  • Zhu FC, Wang H, Fang HH, Yang JG, Lin XJ, Liang XF, Zhang XF, Pan HX, Meng FY, Hu YM, et al. A novel influenza A (H1N1) vaccine in various age groups. N Engl J Med. 2009 Dec 17;361(25):2414–23. doi:10.1056/NEJMoa0908535. PMID:19846844.
  • Monto AS, Malosh RE, Petrie JG, Martin ET. The Doctrine of Original Antigenic Sin: separating Good From Evil. J Infect Dis. 2017 Jun 15;215(12):1782–88. doi:10.1093/infdis/jix173. PMID:28398521.
  • Andrews SF, Kaur K, Pauli NT, Huang M, Huang Y, Wilson PC. High preexisting serological antibody levels correlate with diversification of the influenza vaccine response. J Virol. 2015 Mar;89(6):3308–17. PMID:25589639. doi:10.1128/jvi.02871-14.
  • Sasaki S, He XS, Holmes TH, Dekker CL, Kemble GW, Arvin AM, Greenberg HB. Influence of prior influenza vaccination on antibody and B-cell responses. PloS One. 2008 Aug 20;3(8):e2975. doi:10.1371/journal.pone.0002975. PMID:18714352.
  • Sperling RS, Engel SM, Wallenstein S, Kraus TA, Garrido J, Singh T, Kellerman L, Moran TM. Immunogenicity of trivalent inactivated influenza vaccination received during pregnancy or postpartum. Obstet Gynecol. 2012 Mar;119(3):631–39. PMID:22353963. doi:10.1097/AOG.0b013e318244ed20.
  • Thompson MG, Naleway AL, Fry AM, Ball S, Spencer S, Reynolds S, Bozeman S, Levine MZ, Katz JM, Gaglani M. Effects of Repeated Annual Inactivated Influenza Vaccination among Healthcare Personnel on Serum Hemagglutinin Inhibition Antibody Response to A/Perth/16/2009 (H3N2)-like virus during 2010-11. Vaccine. 2016;34(7):981–88. doi:10.1016/j.vaccine.2015.10.119.
  • Khurana S, Hahn M, Coyle EM, King LR, Lin T, Treanor JJ, Sant AJ, Golding H. Repeat vaccination reduces antibody affinity maturation across different influenza vaccine platforms in humans. Nat Commun. 2019;10(1):3338. doi:10.1038/s41467-019-11296-5.
  • Poland GA, Ovsyannikova IG, Jacobson RM, Smith DI. Heterogeneity in vaccine immune response: the role of immunogenetics and the emerging field of vaccinomics. Clin Pharmacol Ther. 2007 Dec;82(6):653–64. PMID:17971814.doi:10.1038/sj.clpt.6100415.
  • Linnik JE, Egli A. Impact of host genetic polymorphisms on vaccine induced antibody response. Hum Vaccin Immunother . PMID:26809773. 2016 Apr 2;12(4):907–15. doi:10.1080/21645515.2015.1119345.
  • Posteraro B, Pastorino R, Di Giannantonio P, Ianuale C, Amore R, Ricciardi W, Boccia S. The link between genetic variation and variability in vaccine responses: systematic review and meta-analyses. Vaccine. 2014 Mar 26;32(15):1661–69. doi:10.1016/j.vaccine.2014.01.057. PMID:24513009.
  • Mert G, Sengul A, Gul HC, Karakas A, Eyigun CP. The role of human leukocyte antigen tissue groups in hepatitis B virus vaccination in Turkey. Journal of Microbiology, Immunology, and Infection = Wei Mian Yu Gan Ran Za Zhi. 2014 Feb;47(1):9–14. PMID:23523043. doi:10.1016/j.jmii.2013.01.004.
  • Reali G. [The HLA system and the major histocompatibility complex in humans]. Pathologica. 1975 Nov–Dec;67(973–974):439–51. Italian. PMID:131932.
  • Mackenzie JS, Wetherall JD, Fimmel PJ, Hawkins BR, Dawkins RL. Host factors and susceptibility to influenza A infection: the effect of ABO blood groups and HL-A antigens. Dev Biol Stand. Jun1 1977;39:355–62. PMID:604120.
  • Moss AJ, Gaughran FP, Karasu A, Gilbert AS, Mann AJ, Gelder CM, Oxford JS, Stephens HA, Lambkin-Williams R. Correlation between human leukocyte antigen class II alleles and HAI titers detected post-influenza vaccination. PloS One . 2013;8(8):e71376. doi:10.1371/journal.pone.0071376. PMID:23951151.
  • Poland GA, Ovsyannikova IG, Jacobson RM Immunogenetics of seasonal influenza vaccine response. Vaccine. Sep12 2008;26Suppl 4: D35-40. PMID:19230157. doi: 10.1016/j.vaccine.2008.07.065. PMID:19230157.
  • Narwaney KJ, Glanz JM, Norris JM, Fingerlin TE, Hokanson JE, Rewers M, Hambidge SJ. Association of HLA class II genes with clinical hyporesponsiveness to trivalent inactivated influenza vaccine in children. Vaccine. 2013 Feb 4;31(7):1123–28. doi:10.1016/j.vaccine.2012.12.026. PMID:23261040.
  • Schirmer M, Kumar V, Netea MG, Xavier RJ. The causes and consequences of variation in human cytokine production in health. Curr Opin Immunol. 2018 Oct;54:50–58. doi:10.1016/j.coi.2018.05.012. PMID:29913309.
  • Egli A, Santer DM, O’Shea D, Barakat K, Syedbasha M, Vollmer M, Baluch A, Bhat R, Groenendyk J, Joyce MA, et al. IL-28B is a key regulator of B- and T-cell vaccine responses against influenza. PLoS Pathog. 2014 Dec;10(12):e1004556. doi:10.1371/journal.ppat.1004556. PMID:25503988..
  • Ovsyannikova IG, White SJ, Larrabee BR, Grill DE, Jacobson RM, Poland GA. Leptin and leptin-related gene polymorphisms, obesity, and influenza A/H1N1 vaccine-induced immune responses in older individuals. Vaccine. 2014 Feb 7;32(7):881–87. doi:10.1016/j.vaccine.2013.12.009. PMID:24360890.
  • Qin L, Wang D, Li D, Zhao Y, Peng Y, Wellington D, Dai Y, Sun H, Sun J, Liu G, et al. High Level Antibody Response to Pandemic Influenza H1N1/09 Virus Is Associated With Interferon-Induced Transmembrane Protein-3 rs12252-CC in Young Adults. Front Cell Infect Microbiol . 2018;8:134. doi:10.3389/fcimb.2018.00134. PMID:29868492.
  • Lei N, Li Y, Sun Q, Lu J, Zhou J, Li Z, Liu L, Guo J, Qin K, Wang H, et al. IFITM3 affects the level of antibody response after influenza vaccination. Emerging Microbes & Infections. 2020;9(1):976–87. PMID:32321380. doi:10.1080/22221751.2020.1756696.
  • Cummins NW, Weaver EA, May SM, Croatt AJ, Foreman O, Kennedy RB, Poland GA, Barry MA, Nath KA, Badley AD. Heme oxygenase-1 regulates the immune response to influenza virus infection and vaccination in aged mice. Faseb J. 2012;26(7):2911–18. doi:10.1096/fj.11-190017. PMID:22490782.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.