3,458
Views
51
CrossRef citations to date
0
Altmetric
Review

Factors affecting immune responses to the influenza vaccine

Pages 637-646 | Received 01 Mar 2017, Accepted 31 May 2017, Published online: 21 Jul 2017

ABSTRACT

Annual administration of the seasonal influenza vaccine is strongly recommended to reduce the burden of disease, particularly for persons at the highest risk for the viral infection. Even during years when there is a good match between the vaccine and circulating strains, host-related factors such as age, preexisting immunity, genetic polymorphisms, and the presence of chronic underlying conditions may compromise influenza vaccine responsiveness. The application of new methodologies and large-scale profiling technologies are improving the ability to measure vaccine immunogenicity and our understanding of the immune mechanisms by which vaccines induce protective immunity. This review attempts to summarize the general concepts of how host factors can contribute to the heterogeneity of immune responses induced by influenza vaccines.

Introduction

Influenza is a contagious acute respiratory disease that causes seasonal epidemics with 3–5 million hospitalizations and 250,000–500,000 deaths annually worldwide (WHO influenza center website). Although influenza viruses can cause disease in any age group, complications leading to serious illness and death following infection are predominantly observed among those aged ≥ 65 y and immunocompromised individuals. Thus, annual influenza vaccination has long been recommended for these vulnerable and most-at-risk groups.

Currently, licensed influenza vaccines contain components of 3/4 virus strains responsible for seasonal epidemics that are changed annually on the basis of global surveillance data.Citation1 Vaccine-induced neutralizing antibodies against the hemagglutinin (HA) represent the most effective correlate of protection against homologous influenza virus strains.Citation2 Even during years when there is a good match between the vaccine and the circulating strains, influenza vaccine effectiveness (VE) in preventing influenza illness may vary among vaccine recipients depending on age and health status of the individual.Citation3,4 In addition, preexisting immunity and genetic and hormonal factors may account for the inter-individual variability with respect to the immunological parameters used for assessing vaccine responsiveness. Over the last few years, new methodologies have increased our knowledge on the cellular and molecular mechanisms of vaccine immunity and help explain how host factors contribute to mechanisms that mediate the efficacy of vaccines against influenza.

Preexisting immunity

The impact of influenza exposure history from prior infections and vaccinations on current-season strains remains controversial.Citation5-9 However, there are few data related to the effect of vaccinations received in previous seasons, and these data may be affected by bias and uncontrolled confounding. Thus, annual VE studies in large population groups of varying ages and that consider virus type/subtype and the vaccine type would help determine the appropriate response to annual vaccination. Furthermore, the specificity of the end-point or outcome measure used in the study should be considered. Currently, hemagglutination inhibition (HI) titers are used to evaluate immunogenicity of influenza vaccines, and values ≥ 40 are used as the surrogate correlate of 50% protection in adults.Citation10,11 Low rates of seroconversion after a trivalent inactivated influenza vaccine (TIV) administration and low ratios of post- to pre-vaccination HI titers are usually observed in subjects who have been repeatedly vaccinated,Citation12 although some studies have shown that the influenza vaccine was able to raise and maintain protective antibody titers.Citation13,14 In addition, cell-mediated immune responses may correlate better with protection in these individuals than HI titers.Citation15-18

The application of new methodologies has improved our understanding of antibody-mediated responses to vaccination or infection. Andrews et al.Citation19 recently determined how the specificity of B cell responses are biased by recent past exposures. The authors conducted a longitudinal analysis of the plasmablasts, memory and serological responses upon vaccination with TIV in multiple individuals between 2006 and 2013. They found that repeated vaccination with the same vaccine strains from year to year reduced the overall vaccine-induced B cell response. In particular, high preexisting serological antibody levels to an influenza virus strain correlate with a low production of antibody secreting cells and memory B cells following repeated vaccination. Conversely, a robust B cell response is generated from antigenically novel vaccine strains. Preexisting serum antibodies, acquired through multiple immunizations, are hypothesized to bind and mask viral epitopes in the vaccine, thus reducing activation of memory B cells by recognizing these particular epitopes. Thus the level of preexisting serum antibodies modulates the magnitude of the response to repeated vaccinations, ensuring that a broad and diverse memory pool is ready to respond to a wide range of virus variants.

Fonville et al.Citation20 recently introduced the antibody landscape, which is a novel method for determining and representing serum antibody titers toward different antigenic variants in response to influenza infection or vaccination. The authors use antigenic cartographyCitation21 to map the antigenic relationship between influenza strains; then, HI titers of a given serum are added with elevations corresponding to antigenic variants with higher antibody levels. A substantial heterogeneity among the antibody landscapes of 69 individuals monitored for infection over 6 y was observed, although each individual antibody landscape was stable from one year to the next. In another cohort, the in-depth serological phenotyping of 225 individuals who received a previously encountered vaccine strain or a more novel strain showed that the use of an antigenically updated virus variant stimulated antibodies to the novel variant and provided “back boosting” of titers to previous antigenic clusters.Citation20 By using this approach, the authors could quantify this “back boosting” and correlate the antigenic differences among the influenza viruses. Importantly, their findings indicate that vaccine updates may be beneficial and improve vaccine efficacy in previously exposed individuals.

Molecular-based technologies have strongly contributed to provide data describing immune cells and their functions. Lee et al.Citation22 described a molecular-level analysis of the serum antibody repertoire in a relatively small cohort of young adults before and after seasonal influenza vaccination. A high-resolution proteomics analysis of immunoglobulins coupled with high-throughput sequencing of transcripts encoding B cell receptors was used to characterize the antibody repertoire at the individual clonotype level in the sera of these subjects. By these means, the authors could demonstrate that high serum antibody titers before vaccination strongly correlate with the boosting of preexisting antibodies and the emergence of fewer vaccine-elicited antibodies. In particular, they found an unexpectedly high fraction of serum antibodies recognized by both the H1 and H3 monovalent vaccines, thus providing new insights for the development of a universal influenza vaccine.

Even though strain-specific neutralizing antibodies confer protection against infection with matching influenza virus strains, non-neutralizing antibodies and cellular immune responses to the virus that are cross-reactive to other viral strains also contribute to reduce disease severity and infectivity. A meta-analysis of 5 clinical studies performed between 2008 and 2013 in individuals receiving influenza live viral challenges provides further evidence on the ability of preexisting heterosubtypic cellular immunity to reduce both viral shedding and symptom scores in humans.Citation23 Notably, Trieu et al.Citation24 showed that both strain-specific and cross-reactive CD4+ and CD8+ T cells were maintained in healthcare workers who received a single AS03-adjuvanted H1N1pdm09 vaccine in 2009 and subsequently repeated annual vaccinations, whereas these cells declined significantly in those who received a single vaccine and no further vaccination. In addition, recent evidence that inactivated influenza vaccines could induce ADCC-antibodies against both homologous and heterologous influenza viruses in children and adults and that stalk-reactive antibodies appear to be preexistent within the memory compartment further highlights the role of preexisting cross-reactive immune responses against influenza.Citation25,26

Recently, Gostic et al.Citation27 hypothesized that childhood HA imprinting may provide lifelong protection against severe infection and death from HA subtypes in the same phylogenetic group. In particular, their analysis of all known human cases of H5N1 and H7N9 with reported patient age revealed strong evidence that individuals born before the emergence of H3N2 (group 2) in 1968 showed protection against severe cases of H5N1 (group 1) but not H7N9 (group 2), and those born after 1968 showed the opposite pattern. As suggested by the authors, HA group imprinting also may contribute to the greater impact of seasonal H3N2 compared with H1N1 in older age groups. These findings provide new insights on herd immunity against zoonotic influenza virus strains in the human population and raise questions about the impact of influenza vaccination both on imprinting and boosting cross-reactive anti-HA responses that might be relevant for future research.

Overall, influenza exposure history based on prior infections and vaccinations heavily contributes to the complexity of the polyclonal antibody response to influenza viral strains in humans. For this reason, multiple approaches are necessary and may help explain the heterogeneity found among several serological studies. In addition, a better understanding of the impact of cross-reactive immune responses to influenza protection would help determine possible additional benefits provided by repeat vaccines.

Immunosenescence

Over the past years, several studies have been conducted with large numbers of individuals and across multiple influenza seasons to assess vaccine efficacy and effectiveness in older adults. Although sometimes controversial when comparing older adults to unvaccinated individuals, the results clearly show that vaccine responses in older adults are reduced compared with young adults.Citation28,29 Indeed, influenza vaccines are estimated to be < 50% effective against influenza in older individuals even with a high rate of influenza vaccination and a well-matched vaccine.Citation3,30-32

Physiological changes and a decline in immune function termed “immunosenescence” are responsible for a higher susceptibility to influenza, higher mortality rates and a poor influenza vaccine response. The influence of this age-related dysfunction on the innate and adaptive immune systems has been largely documented.Citation33,34 In particular, inappropriate activation of Toll-like receptors (TLRs) and costimulatory molecules in immune cells from older adults directly affects both cellular and humoral immunity and thus influenza-vaccine-induced antibody production.Citation35 The function and number of NK cells are altered in individuals with a diminished health status and correlate with low HI titers in vaccinated older individuals.Citation36,37 In addition, defects in the generation and function of B and T cells, reduced production of high affinity antibodies, decreased expression of the costimulatory molecule CD28 on T cells, reduced telomerase activity, and Th1/Th2 cytokine disbalance were observed with aging, and parameters related to these functions are considered immunosenescence markers.Citation38-43 Furthermore, aging is characterized by a low-grade chronic inflammatory status, called “inflammaging,” which is measured by circulating levels of IL-6, TNF-α and C-reactive protein, and latent infections with viruses such as cytomegalovirus (CMV).Citation44,45 In particular, latent CMV infection has been associated with an accelerated aging of the immune system in the elderly, and the virus has a negative effect on the in vivo and in vitro B cell responses to the seasonal influenza vaccine.Citation46,47 Recently, Frasca et al.Citation48 demonstrated for the first time a negative association between CMV seropositivity and the B cell predictive biomarkers of effective vaccine responses such as switched memory B cells and activation-induced cytidine deaminase in CpG-stimulated B cell cultures. In addition, CMV may also down-regulate the influenza vaccine-specific antibody response through the induction of late-differentiated T cells and the accumulation of senescent T cells, which lead to reduced influenza-specific memory T cells.Citation46,49 Thus, many components of the immune system contribute to the ability of an older individual to mount an effective response following vaccination against influenza. Because of this, a systems biology generated immune profile pre- and post-vaccination is essential to provide insight into the multiple signaling pathways and immunological processes.

Systems biology approaches have been used to identify molecular signatures of vaccine-induced immune responses in humans.Citation50-52 Among these, an increased expression of TNFRS17, which encodes the BCMA receptor for the B cell growth factor BAFF implicated in B cell differentiation, was a predictor of the antibody response in healthy adults vaccinated with TIV.Citation53 By integrating transcriptomic and microRNA (miRNA) expression data from a longitudinal study across 5 influenza seasons in diverse populations, Nakaya et al.Citation54 were able to identify several potential miRNA regulators of the interferon response to TIV vaccination. Importantly, their results revealed a negative association between enhanced NK cell and monocyte numbers in the elderly compared with young subjects and the magnitude of antibody response to vaccination. Furthermore, interferon-regulatory transcription factors (IRF1/IRF2/IRF6/IRF7/IRF9), chemokine/chemokine receptors (CCR5/CCR9/CCL5), cytokine/cytokine receptors (IFNG/IL10RA/TNFRSF1A), protein kinases (MAP2K4/MAPK3), growth factor receptor (TGFBR1) and several uncharacterized genes, such as ZNF300, NUP1333 and KLK1, were associated with the antibody response following influenza A/H1N1 vaccination in older adults.Citation55 Poland and collaborators, who first coined the term vaccinomics, in which information across transcriptomic, genomic, and proteomic data can be correlated with humoral and cellular immune measures to gain a greater understanding on vaccine immunogenicity, have also associated epigenetic markers with the influenza vaccine response.Citation56,57 In a recent work,Citation58 they show that sites of methylation regulation associated with the humoral response to vaccination in a cohort of 158 50-to 74-year-old individuals affect known cellular differentiation signaling and antigen presentation pathways. Specifically, they identified a group of CpGs that, when coordinately methylated, are associated with a higher antibody response, and when hypo-methylated are associated with a lower response. The availability of data sets (DNA methylation, mRNA expression, miRNA expression, and proteomics) could also provide evidence that, in addition to dysregulated immune function, immunosenescence may have effects on underlying biological processes and a variety of metabolic activities.Citation59

Age-dependent comorbidities, including metabolic disorders, may also have an effect on the quality of the immune response to the influenza vaccine. Dementia and undernutrition, which are very common in frail elderly patients in geriatric medical long-term care, have been frequently observed among poor responders in vaccination studies.Citation60-63 The availability of high-dose influenza vaccines, intradermal influenza vaccines and MF59-adjuvanted vaccines has increased immunogenicity for influenza antigens in older adults.Citation64-66 Nonetheless, the immune responses in these individuals never achieve the levels observed in young adults receiving a standard vaccination.Citation67 Advances in scientific methodology and supporting technology over the years have contributed to increasing our knowledge in the field and may help improve current vaccination for the older adult population. Increasing evidence suggests that the modulation of signal transduction pathways such as the mammalian target of rapamycin (mTOR) pathway may have beneficial effects on aging and age-related conditions in humans. In particular, Mannick et al.Citation68 found that a 6-week treatment with the mTOR inhibitor RAD001, an analog of rapamycin, improved the response to influenza vaccination in elderly volunteers ≥ 65 y of age. A decrease in the percentage of PD-1-positive T cells observed in RAD001-treated cohorts compared with the placebo cohort may likely contribute to enhanced immune function in the elderly. Furthermore, RAD001 treatment increased the serologic cross-reactive response to heterologous strains of influenza not contained in the seasonal influenza vaccine. Keating et al.Citation69 also reported that treatment with rapamycin during immunization could provide cross-protective immunity against lethal infections with the influenza virus in mice. Rapamycin reduced the formation of germinal centers and inhibited class switching in B cells, facilitating the production of antibodies of lower affinity with a greater potential to be cross-reactive. In summary, these studies show how insights into the pathways may help guide the development of novel vaccines and strategies that provide better protection of at-risk populations in the future.

Genetic polymorphism, sex-based differences and obesity

Several studies document the influence of the host genetic background on the immune response to influenza vaccination.Citation70-72 Polymorphisms of any of the involved genes in the molecular interactions within the immune system reflect the high variability between individuals to respond efficiently to vaccines. Even in healthy young individuals, there are subjects who do not develop a protective immune response to the influenza vaccine.

Associations between HLA class II alleles and nonresponsiveness to influenza vaccination were clearly observed in individuals with HLA-DRB1*07, who produced lower antibody titers following a single administration of the influenza vaccine.Citation73 Conversely, a lower frequency of HLA-DBQ1*0603–9/14 (and DRB1*13) was found in nonresponders, compared with matched responders receiving the same vaccine.Citation73 A further study performed in an elderly cohort of recipients showed that HLA-DRB1*04:01 and HLA-DPB1*04:01 occurred at higher frequencies in individuals with seroprotective levels of the anti-HA antibodies compared with those non-seroprotected following influenza vaccination.Citation74

Single-genetic polymorphisms (SNPs) in other genes have been associated with the influenza-specific antibody response after vaccination. Poland et al.Citation75 found associations of several SNPs in coding and noncoding regions of cytokine (IL6, IL18, IL12A IL12B, IFNg) and cytokine receptor (IL1R, IL2RG, IL4R, IL10RB, IL12RB, IFNAR2, TNFRS F1A) genes and H1N1 antibody titers in 184 18-to 40- year-old healthy recipients of the seasonal influenza vaccine. A recent work by Egli et al.Citation76 shows that a SNP in IL28B, a member of the IFN-λ family, could be associated with an increased seroconversion rate after influenza vaccination. Notably, by using antagonist peptides to block the IL28 receptor subunit (IL28RA) in vitro, the authors could identify IL28B as a key regulator of the Th1/Th2 balance in influenza vaccination. Furthermore, Franco et al.Citation77 identified 20 genes involved in generalized biological functions, such as intracellular transport and membrane trafficking, that contributed to differential immune responses of healthy adults to influenza vaccination. Overall, the large number of SNPs that may be involved in immune responses highlights the complex interactions between different cell types and the need for a multidisciplinary approach with evidence-based principles that may be adopted for predicting influenza vaccine responsiveness.

Although limited data exist, sex-differences have also been reported in response to diverse vaccines across different age groups.Citation78,79 Both younger and older females have greater antibody responses than males following seasonal TIV. Engler et al.Citation80 also reported that healthy women who were vaccinated with a half dose of TIV had antibody titers similar to those in men who were given a full dose. Among older adults who received the high-dose TIV, Falsey et al.Citation64 documented higher antibody responses against each of the 3 influenza antigens in females compared with males. Sex steroid hormones contribute to differences in both innate and immune responses to infection and vaccination in men and women by directly binding to intracellular receptors in immune cells such as monocytes, B cells and T cells and the subsequent activation of hormone-responsive genes.Citation79,81 However, the age-related reduction of estrogen levels in women partially alters the sex-bias in vaccination. Conversely, there is evidence for an immunosuppressive role of testosterone in the response to influenza vaccination. Furman et al.Citation82 used a systems approach for the analysis of sex differences in the immune system and suggested that the natural variation in circulating free testosterone could explain the differences observed in the response to vaccines. They found that men with elevated serum testosterone levels and a high expression of genes involved in lipid metabolism had the lowest antibody responses to TIV. By repressing transcription factors such as FOS and JUN, which are involved in immune activation, testosterone would repress their further function to modulate genes implicated in the metabolism of lipids. In addition to sex hormones, other genes located on the X chromosome and encoding proteins such as TLR7, TLR8, GATA1, IRAK1, CD40L and FOXP3 may affect the immune response to vaccination.Citation83 Furthermore, the prevalence of miRNAs, which are crucial regulators of gene expression, on the X chromosome compared with the low number contained in the Y chromosome strongly implicate a role in sex-based differences in immunity.Citation84

Obesity results in ineffective immune responses to influenza virus infection and represents a relevant risk factor for increased severity of influenza symptoms.Citation85-87 Paich et al.Citation88 first indicated that CD4+ and CD8+ T cells from obese individuals have substantial defects in activation and function that likely contribute to the increased morbidity and mortality from the H1N1pdm09 virus observed in these individuals compared with healthy weight adults. Obesity has also been clearly associated with a decreased immune response to influenza vaccination. Previous studies performed in mice subjected to diet-induced obesity showed a marked suppression of antibody production and neutralizing activity in response to a H1N1pdm09 vaccine, and thus a dramatically reduced protective efficacy against H1N1 virus challenge, compared with lean control mice.Citation89,90 A recent workCitation91 showed that despite the increased seroconversion conferred by the AS03-adjuvanted H1N1pdm09 vaccine, obese mice still succumbed to the influenza virus challenge and had higher morbidity and mortality compared with normal-weight mice. Human studies have had various results. Sheridan et al.Citation92 reported, for the first time, that a higher initial fold increase in antibodies to TIV in healthy obese individuals was followed, 12 months post-vaccination, by a greater decline in antibody titers and defective CD8+ T cell responses compared with healthy weight individuals. Recently, Frasca et al.Citation93 found reduced antibody responses to influenza vaccination in both young and elderly obese individuals that was associated with a decreased B cell function and higher levels of intracellular TNF-α and TLR4 compared with those detected in lean individuals. By contrast, other studies reported that antibody responses to influenza vaccination were similar in obese and lean older adults and among children and adolescents of various body mass index.Citation94,95 Recently, Esposito et al.Citation96 also found that in overweight and obese children aged 3–14 years, antibody responses to TIV were similar or in some cases slightly higher than in lean subjects of a similar age, even 4 months after vaccination. In addition to these conflicting results, obesity is a complex clinical condition that impairs various functions. Even if seroprotective antibodies are elicited, vaccination efficacy in obese individuals may be affected by obesity-associated decreases in key immunological and biological functions critical to counteract an influenza virus infection. However, this increasing population worldwide at a high risk of infection would have benefits from vaccination and thus should regularly receive a seasonal influenza vaccine.

Presence of chronic underlying medical conditions

Individuals with autoimmune inflammatory diseases, such as rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE), or cancer are at an increased risk of influenza virus infection because of their disease-associated impaired imbalances and the use of immunosuppressive therapies. Reduced immunogenicity of the influenza vaccine appears to reflect the impaired immunity in these patients, and the variability in the results of some studies are likely related to several factors, including disease heterogeneity and the varying degree of the patients' lymphopenia.Citation97-100 Nevertheless, there is evidence of influenza vaccine efficacy and effectiveness in these immunocompromised individuals.Citation101-104 In particular, Liao et al.Citation105 conducted a meta-analysis of influenza vaccine seroprotection, seroconversion and adverse effects in SLE patients. In this context, 18 studies with 1966 subjects, of whom at least 565 were patients with low-to-moderate SLE Disease Activity Index scores or SLE disease, showed that drugs such as glucocorticoids, azathioprine, methotrexate and mycophenolate mofetil decreased humoral responses to influenza vaccination compared with healthy controls. However, the immunogenicity of the influenza vaccine in SLE patients almost met that of the “Committee for Proprietary Medicinal Products” (CPMP) guidelines.Citation106 The mild and manageable side effects, detected in some of the SLE patients, further support the beneficial effects of influenza vaccination with TIV.

The current recommendation for cancer patients is to receive the influenza vaccine before the start of cytotoxic chemotherapy.Citation107-109 However, Keam et al.Citation110 conducted a large randomized clinical trial to determine the optimal timing of influenza vaccination during 3-week cytotoxic chemotherapy cycles. In particular, 83 adult patients with breast or lung cancer were randomly divided into 2 subgroups to receive the 2014–2015 seasonal TIV on day 1 or 11 during the chemotherapy cycle, and then stratified by age (< 60 and >60 years) and prior influenza vaccination status. Although generally lower than in healthy adults, antibody responses against influenza vaccine strains were comparable for patients who received the vaccine concurrently with chemotherapy (day 1) or within the cytopenic period (day 11).

In HIV-infected patients, depletion of CD4+ T cells due to HIV infection causes immunodeficiency and thus a higher risk of developing influenza-related complications. Despite a reduced immune response, the influenza vaccine appears to be safe and effective in reducing influenza disease in these patients.Citation111,112In particular, early control of viremia by antiretroviral treatment (ART) can partially reverse B cell dysregulation and preserve influenza vaccine responsiveness.Citation113-115Recently, Rinaldi et al.Citation116measured both antibody and memory B-cell responses to the trivalent 2012–13 seasonal influenza vaccine in a cohort of vertically HIV-1 infected children and young adults receiving an influenza vaccination annually. Despite lower seroconversion rates after vaccination compared with healthy controls, similar frequencies of influenza-specific memory B cells for the 3 vaccine strains were detected by B-cell ELISPOT in both groups, suggesting that qualitative measures are more informative than standard serological markers in this population. Somewhat different results were reported by Wheatley et al.Citation117 who found lower frequencies and thus impaired induction of memory B cells in HIV+ individuals to influenza vaccination by using direct staining of HA-specific B cells with recombinant HA probes. Differences between the 2 studies may be due to the different cohorts analyzed or alternative methodologies used for memory B-cell measurements.

Transplant recipients, individuals with chronic obstructive pulmonary disease, and individuals with Down syndrome also have impaired immune responses to vaccination, including influenza.Citation118-120 Collectively, there is a need for more in-depth studies aimed at identifying the appropriate immunologic parameters to evaluate immunogenicity and assess vaccine effectiveness in immunocompromised individuals.

Conclusions

Several host-related factors such as age, genetic polymorphisms, and the presence of chronic underlying conditions may contribute to a decline in immune responses and consequently compromise influenza vaccine responsiveness. Although complex and still challenging in many aspects, high-throughput technologies and the analysis of multi-omics experiments hold great promise to characterize the interactions between individual components of the immune system and thus better understand and predict vaccine-induced immunity.Citation121,122 Because of these studies, it is reasonable to believe that new insights about the efficacy of TIV in special populations such as the elderly or immunocompromised may enable rational design of specific vaccine formulations that target these most-at-risk groups.

In the meantime, further studies of influenza vaccination using alternative criteria rather than HI seroconversion to categorize subjects into ‘responders” and ‘nonresponders’ will certainly help to establish the influenza vaccine efficacy. Among these, the use of human challenge models would be extremely informative for correlating the protection and development of new drugs and vaccines.Citation123,124In addition, there are intense ongoing efforts to develop more immunogenic and broadly protective influenza vaccines that are based on highly conserved viral antigens, such as the HA stalk region and internal viral proteins, and early results seem to be promising.Citation125,126

Lastly, and more importantly, the efficacy of current licensed vaccines may be lower in high-risk groups than in normal subjects, but the vaccines are still capable of reducing disease severity and mortality. Although countries have recommendations in place, vaccination coverage rates remain too low, and there is a need to improve vaccine awareness and education among patients, their families and healthcare workers to prevent the hospitalizations and deaths attributed to influenza in these groups each year.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Funding

This work was supported by the Ministry of Health, Italy (grant number RF-2010–2318269).

References

  • Grohskopf LA, Sokolow LZ, Broder KR, Olsen SJ, Karron RA, Jernigan DB, Bresee JS. Prevention and control of seasonal influenza with vaccines. MMWR Recomm Rep 2016; 65:1-54; PMID:27560619; https://doi.org/10.15585/mmwr.rr6505a1
  • Seidman JC, Richard SA, Viboud C, Miller MA. Quantitative review of antibody response to inactivated seasonal influenza vaccines. Influenza Other Respir Viruses 2012; 6:52-62; PMID:21668661; https://doi.org/10.1111/j.1750-2659.2011.00268.x
  • Osterholm MT, Kelley NS, Sommer A, Belongia EA. Efficacy and effectiveness of influenza vaccines: a systematic review and meta-analysis. Lancet Infect Dis 2012; 12:36-44; PMID:22032844; https://doi.org/10.1016/S1473-3099(11)70295-X
  • Domínguez A, Godoy P, Torner N. The Effectiveness of Influenza Vaccination in Different Groups. Expert Rev Vaccines 2016; 15:751-64; PMID:26775669; https://doi.org/10.1586/14760584.2016.1142878
  • Beyer WE, de Bruijn IA, Palache AM, Westendorp RG, Osterhaus AD. Protection against influenza after annually repeated vaccination: a meta-analysis of serologic and field studies. Arch Intern Med 1999; 159:182-8; PMID:9927102; https://doi.org/10.1001/archinte.159.2.182
  • Keitel WA, Cate TR, Couch RB, Huggins LL, Hess KR. Efficacy of repeated annual immunization with inactivated influenza virus vaccines over a five year period. Vaccine 1997; 15:1114-22; PMID:9269055; https://doi.org/10.1016/S0264-410X(97)00003-0
  • Sullivan SG, Kelly H. Stratified estimates of influenza vaccine effectiveness by prior vaccination: caution required. Clin Infect Dis 2013; 57:474-6; PMID:23619811; https://doi.org/10.1093/cid/cit255
  • Ohmit SE, Thompson MG, Petrie JG, Thaker SN, Jackson ML, Belongia EA, Zimmerman RK, Gaglani M, Lamerato L, Spencer SM, et al. Influenza vaccine effectiveness in the 2011–2012 season: protection against each circulating virus and the effect of prior vaccination on estimates. Clin Infect Dis 2014; 58:319-27; PMID:24235265; https://doi.org/10.1093/cid/cit736
  • McLean HQ, Thompson MG, Sundaram ME, Meece JK, McClure DL, Friedrich TC, Belongia EA. Impact of repeated vaccination on vaccine effectiveness against influenza A(H3N2) and B during 8 seasons. Clin Infect Dis 2014; 59:1375-85; PMID:25270645; https://doi.org/10.1093/cid/ciu680
  • Nauta JJ, Beyer WE, Osterhaus AD. On the relationship between mean antibody level, seroprotection and clinical protection from influenza. Biologicals 2009; 37:216-21; PMID:19268607; https://doi.org/10.1016/j.biologicals.2009.02.002
  • Coudeville L, Bailleux F, Riche B, Megas F, Andre P, Ecochard R. Relationship between haemagglutination-inhibiting antibody titres and clinical protection against influenza: development and application of a bayesian random-effects model. BMC Med Res Methodol 2010; 10:18; PMID:20210985; https://doi.org/10.1186/1471-2288-10-18
  • Thompson MG, Naleway A, Fry AM, Ball S, Spencer AM, Reynolds S, Bozeman S, Levine M, 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; 34:981-8; PMID:26813801; https://doi.org/10.1016/j.vaccine.2015.10.119
  • Kunzel W, Glathe H, Engelmann H, Van Hoecke C. Kinetics of humoral antibody response to trivalent inactivated split influenza vaccine in subjects previously vaccinated or vaccinated for the first time. Vaccine 1996; 14:1108-10; PMID:8911005; https://doi.org/10.1016/0264-410X(96)00061-8
  • De Bruijn IA, Remarque EJ, Jol-van der Zijde CM, van Tol MJ, Westendorp GG, Knook DL. Quality and quantity of the humoral immune response in healthy elderly and young subjects after annually repeated influenza vaccination. J Infect Dis 1999; 179:31-6; PMID:9841819; https://doi.org/10.1086/314540
  • McElhaney JE, Xie D, Hager WD, Barry MB, Wang Y, Kleppinger A, Ewen C, Kane KP, Bleackley RC. T cell responses are better correlates of vaccine protection in the elderly. J Immunol 2006; 176:6333-9; PMID:16670345; https://doi.org/10.4049/jimmunol.176.10.6333
  • McElhaney JE, Xie D, Hager WD, Barry MB, Wang Y, Kleppinger A, Ewen C, Kane KP, Bleackley RC. Granzyme B: correlates with protection and enhanced CTL response to influenza vaccination in older adults. Vaccine 2009; 27:2418-25; PMID:19368783; https://doi.org/10.1016/j.vaccine.2009.01.136
  • Zeman AM, Holmes TH, Stamatis S, Tu W, He XS, Bouvier N, Kemble G, Greenberg HB, Lewis DB, Arvin AM, Dekker CL. Humoral and cellular immune responses in children given annual immunization with trivalent inactivated influenza vaccine. Pediatr Infect Dis J 2007; 26:107-15; PMID:17259871; https://doi.org/10.1097/01.inf.0000253251.03785.9b
  • Reber AJ, Kim JH, Biber R, Talbot HK, Coleman AL, Chirkova T, Gross FL, Steward-Clark E, Cao W, Jefferson S, et al. Preexisting immunity, more than aging, influences influenza vaccine responses. Open Forum Infect Dis 2015; 2(2):ofv052; PMID:26380344; https://doi.org/10.1093/ofid/ofv052
  • 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; 89:3308-17; PMID:25589639; https://doi.org/10.1128/JVI.02871-14
  • Fonville JM, Wilks SH, James SL, Fox A, Ventresca M, Aban M, Xue L, Jones TC, Le NM, Pham QT, et al. Antibody landscapes after influenza virus infection or vaccination. Science 2014; 346:996-1000; PMID:25414313; https://doi.org/10.1126/science.1256427
  • Smith DJ, Lapedes AS, de Jong JC, Bestebroer TM, Rimmelzwaan GF, Osterhaus AD, Fouchier RA. Mapping the antigenic and genetic evolution of influenza virus. Science 2004; 305:371-6; PMID:15218094; https://doi.org/10.1126/science.1097211
  • Lee J, Boutz DR, Chromikova V, Joyce MG, Vollmers C, Leung K, Horton AP, DeKosky BJ, Lee CH, Lavinder JJ, et al. Molecular-level analysis of the serum antibody repertoire in young adults before and after seasonal influenza vaccination. Nat Med 2016; 22:1456-64; PMID:27820605; https://doi.org/10.1038/nm.4224
  • Pleguezuelos O, Robinson S, Fernandez A, Stoloff GA, Caparrós-Wanderley W. Meta-analysis and potential role of preexisting heterosubtypic cellular immunity based on variations in disease severity outcomes for influenza live viral challenges in humans. Clin Vaccine Immunol 2015; 22:949-56; PMID:26084515; https://doi.org/10.1128/CVI.00101-15
  • Trieu MC, Zhou F, Lartey S, Jul-Larsen Å, Mjaaland S, Sridhar S, Cox RJ. Long-term maintenance of the influenza-specific cross-reactive memory CD4+ T-cell responses following repeated annual influenza vaccination. J Infect Dis 2017; 215:740-49; PMID:28007925
  • Jegaskanda S, Luke C, Hickman HD, Sangster MY, Wieland-Alter WF, McBride JM, Yewdell JW, Wright PF, Treanor J, Rosenberger CM, Subbarao K. Generation and protective ability of influenza virus-specific antibody-dependent cellular cytotoxicity in humans elicited by vaccination, natural infection, and experimental challenge. J Infect Dis 2016; 214:945-52; PMID:27354365; https://doi.org/10.1093/infdis/jiw262
  • de Vries RD, Nieuwkoop NJ, Pronk M, de Bruin E, Leroux-Roels G, Huijskens EG, van Binnendijk RS, Krammer F, Koopmans MP, Rimmelzwaan GF. Influenza virus-specific antibody dependent cellular cytoxicity induced by vaccination or natural infection. Vaccine 2017; 35:238-47; PMID:27914742; https://doi.org/10.1016/j.vaccine.2016.11.082
  • Gostic KM, Ambrose M, Worobey M, Lloyd-Smith JO. Potent protection against H5N1 and H7N9 influenza via childhood hemagglutinin imprinting. Science 2016; 354:722-26; PMID:27846599; https://doi.org/10.1126/science.aag1322
  • Goodwin K, Viboud C, Simonsen L. Antibody response to influenza vaccination in the elderly: a quantitative review. Vaccine 2006; 24:1159-69; PMID:16213065; https://doi.org/10.1016/j.vaccine.2005.08.105
  • Sasaki S, Sullivan M, Narvaez CF, Holmes TH, Furman D, Zheng NY, Nishtala M, Wrammert J, Smith K, James JA, et al. Limited efficacy of inactivated influenza vaccine in elderly individuals is associated with decreased production of vaccine-specific antibodies. J Clin Invest 2011; 121:3109-19; PMID:21785218; https://doi.org/10.1172/JCI57834
  • Govaert TM, Thijs CT, Masurel N, Sprenger MJ, Dinant GJ, Knottnerus JA. The efficacy of influenza vaccination in elderly individuals. A randomized double-blind placebo-controlled trial. JAMA 1994; 272:1661-5; PMID:7966893; https://doi.org/10.1001/jama.1994.03520210045030
  • Jefferson T, Rivetti D, Rivetti A, Rudin M, Di Pietrantonj C, Demicheli V. Efficacy and effectiveness of influenza vaccines in elderly people: a systematic review. Lancet 2005; 366:1165-74; PMID:16198765; https://doi.org/10.1016/S0140-6736(05)67339-4
  • Beyer WE, McElhaney J, Smith DJ, Monto AS, Nguyen-Van-Tam JS, Osterhaus AD. Cochrane re-arranged: support for policies to vaccinate elderly people against influenza. Vaccine 2013; 31:6030-3; PMID:24095882; https://doi.org/10.1016/j.vaccine.2013.09.063
  • Reber AJ, Chirkova T, Kim JH, Cao W, Biber R, Shay DK, Sambhara S. Immunosenescence and challenges of vaccination against influenza in the aging population. Aging Dis 2012; 3:68-90; PMID:22500272
  • Pinti M, Appay V, Campisi J, Frasca D, Fülöp T, Sauce D, Larbi A, Weinberger B, Cossarizza A. Aging of the immune system: Focus on inflammation and vaccination. Eur J Immunol 2016; 46:2286-301; PMID:27595500; https://doi.org/10.1002/eji.201546178
  • 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. J Immunol 2010; 184:2518-27; PMID:20100933; https://doi.org/10.4049/jimmunol.0901022
  • Mysliwska J, Trzonkowski P, Szmit E, Brydak LB, Machala M, Mysliwski A. Immunomodulating effect of influenza vaccination in the elderly differing in health status. Exp Gerontol 2004; 39:1447-58; PMID:15501014; https://doi.org/10.1016/j.exger.2004.08.005
  • Zhang Y, Wallace DL, de Lara CM, Ghattas H, Asquith B, Worth A, Griffin GE, Taylor GP, Tough DF, Beverley PC, et al. In vivo kinetics of human natural killer cells: the effects of ageing and acute and chronic viral infection. Immunology 2007; 121:258-65; PMID:17346281; https://doi.org/10.1111/j.1365-2567.2007.02573.x
  • Goronzy JJ, Fulbright JW, Crowson CS, Poland GA, O'Fallon WM, Weyand CM. Value of immunological markers in predicting responsiveness to influenza vaccination in elderly individuals. J Virol 2001; 75(24):12182-87; PMID:11711609; https://doi.org/10.1128/JVI.75.24.12182-12187.2001
  • Saurwein-Teissl M, Lung TL, Marx F, Gschösser C, Asch E, Blasko I, Parson W, Böck G, Schönitzer D, Trannoy E, Grubeck-Loebenstein B. 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 2002; 168:5893-9; PMID:12023394; https://doi.org/10.4049/jimmunol.168.11.5893
  • Haynes L, Eaton SM. The effect of age on the cognate function of CD4+ T cells. The frequencies of CD8+ CD28− T cells can be used to predict influenza vaccine response in elderly people. Immunol Rev 2005; 205:220-8; PMID:15882356; https://doi.org/10.1111/j.0105-2896.2005.00255.x
  • Allman D, Miller JP. B cell development and receptor diversity during aging. Curr Opin Immunol 2005; 17:463-7; PMID:16054808; https://doi.org/10.1016/j.coi.2005.07.002
  • Frasca D, Riley RL, Blomberg BB. Humoral immune response and B-cell functions including immunoglobulin class switch are downregulated in aged mice and humans. Semin Immunol 2005; 17:378-84; PMID:15996480; https://doi.org/10.1016/j.smim.2005.05.005
  • Najarro K, Nguyen H, Chen G, Xu M, Alcorta S, Yao X, Zukley L, Metter EJ, Truong T, Lin Y, et al. Telomere length as an indicator of the robustness of B- and T-cell response to influenza in older adults. J Infect Dis 2015; 212:1261-9; PMID:25828247; https://doi.org/10.1093/infdis/jiv202
  • Franceschi C, Bonafè M, Valensin S, Olivieri F, De Luca M, Ottaviani E, De Benedictis G. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci 2000; 908:244-54; PMID:10911963; https://doi.org/10.1111/j.1749-6632.2000.tb06651.x
  • McElhaney JE, Zhou X, Talbot HK, Soethout E, Bleackley RC, Granville DJ, Pawelec G. The unmet need in the elderly: how immunosenescence, CMV infection, co-morbidities and frailty are a challenge for the development of more effective influenza vaccines. Vaccine 2012; 30:2060-7; PMID:22289511; https://doi.org/10.1016/j.vaccine.2012.01.015
  • Trzonkowski P, Myśliwska J, Szmit E, Wieckiewicz J, Lukaszuk K, Brydak LB, Machała M, Myśliwski A. Association between cytomegalovirus infection, enhanced proinflammatory response and low level of anti-hemagglutinins during the anti-influenza vaccination–an impact of immunosenescence. Vaccine 2003; 21:3826-36; PMID:12922116; https://doi.org/10.1016/S0264-410X(03)00309-8
  • Sansoni P, Vescovini R, Fagnoni FF, Akbar A, Arens R, Chiu YL, Cičin-Šain L, Dechanet-Merville J, Derhovanessian E, Ferrando-Martinez S, et al. New advances in CMV and immunosenescence. Exp Gerontol 2014; 55:54-62; PMID:24703889; https://doi.org/10.1016/j.exger.2014.03.020
  • Frasca D, Diaz A, Romero M, Landin AM, Blomberg BB. Cytomegalovirus (CMV) seropositivity decreases B cell responses to the influenza vaccine. Vaccine 2015; 33(12):1433-9; PMID:25659271; https://doi.org/10.1016/j.vaccine.2015.01.071
  • Derhovanessian E, Theeten H, Hähnel K, Van Damme P, Cools N, Pawelec G. Cytomegalovirus-associated accumulation of late-differentiated CD4 T-cells correlates with poor humoral response to influenza vaccination. Vaccine 2013; 31:685-90; PMID:23196209; https://doi.org/10.1016/j.vaccine.2012.11.041
  • Obermoser G, Presnell S, Domico K, Xu H, Wang Y, Anguiano E, Thompson-Snipes L, Ranganathan R, Zeitner B, Bjork A, et al. Systems scale interactive exploration reveals quantitative and qualitative differences in response to influenza and pneumococcal vaccines. Immunity 2013; 38:831-44; PMID:23601689; https://doi.org/10.1016/j.immuni.2012.12.008
  • Tsang JS, Schwartzberg PL, Kotliarov Y, Biancotto A, Xie Z, Germain RN, Wang E, Olnes MJ, Narayanan M, Golding H, et al. Global analyses of human immune variation reveal baseline predictors of postvaccination responses. Cell 2014; 157:499-513; PMID:24725414; https://doi.org/10.1016/j.cell.2014.03.031
  • Li S, Rouphael N, Duraisingham S, Romero-Steiner S, Presnell S, Davis C, Schmidt DS, Johnson SE, Milton A, Rajam G, et al. Molecular signatures of antibody responses derived from a systems biology study of five human vaccines. Nat Immunol 2014; 15:195-204; PMID:24336226; https://doi.org/10.1038/ni.2789
  • Nakaya HI, Wrammert J, Lee EK, Racioppi L, Kunze SM, Haining WN, Means AR, Kasturi N, Khan N, Li GM, et al. Systems biology of vaccination for seasonal influenza in humans. Nat Immunol 2011; 12:786-95; PMID:21743478; https://doi.org/10.1038/ni.2067
  • Nakaya HI, Hagan T, Duraisingham SS, Lee EK, Kwissa M, Rouphael N, Frasca D, Gersten M, Mehta AK, Gaujoux R, et al. Systems Analysis of Immunity to Influenza Vaccination across Multiple Years and in Diverse Populations Reveals Shared Molecular Signatures. Immunity 2015; 43:1186-98; PMID:26682988; https://doi.org/10.1016/j.immuni.2015.11.012
  • Ovsyannikova IG, Oberg AL, Kennedy RB, Zimmermann MT, Haralambieva IH, Goergen KM, Grill DE, Poland GA. Gene signatures related to HAI response following influenza A/H1N1 vaccine in older individuals. Heliyon 2016; 2:e00098; PMID:27441275; https://doi.org/10.1016/j.heliyon.2016.e00098
  • Poland GA, Kennedy RB, Ovsyannikova IG. Vaccinomics and personalized vaccinology: is science leading us toward a new path of directed vaccine development and discovery? PLoS Pathog 2011; 7(12):e1002344
  • Haralambieva IH, Painter SD, Kennedy RB, Ovsyannikova IG, Lambert ND, Goergen KM, Oberg AL, Poland GA. The impact of immunosenescence on humoral immune response variation after influenza A/H1N1 vaccination in older subjects. PLoS One 2015; 10(3):e0122282; PMID:25816015; https://doi.org/10.1371/journal.pone.0122282
  • Zimmermann MT, Oberg AL, Grill DE, Ovsyannikova IG, Haralambieva IH, Kennedy RB, Poland GA. System-wide associations between DNA-methylation, gene expression, and humoral immune response to influenza vaccination. PLoS One 2016; 11(3):e0152034; PMID:27031986; https://doi.org/10.1371/journal.pone.0152034
  • Kennedy RB, Ovsyannikova IG, Haralambieva IH, Oberg AL, Zimmermann MT, Grill DE, Poland GA. Immunosenescence-related transcriptomic and immunologic changes in older individuals following influenza vaccination. Front Immunol 2016; 7:450; PMID:27853459; https://doi.org/10.3389/fimmu.2016.00450
  • Kiecolt-Glaser JK, Glaser R, Gravenstein S, Malarkey WB, Sheridan J. Chronic stress alters the immune response to influenza virus vaccine in older adults. Proc Natl Acad Sci USA 1996; 93:3043-7; PMID:8610165; https://doi.org/10.1073/pnas.93.7.3043
  • Remarque EJ, Cools HJ, Boere TJ, van der Klis RJ, Masurel N, Ligthart GJ. Functional disability and antibody response to influenza vaccine in elderly patients in a Dutch nursing home. BMJ 1996; 312:1015; PMID:8616350; https://doi.org/10.1136/bmj.312.7037.1015
  • Vedhara K, Cox NK, Wilcock GK, Perks P, Hunt M, Anderson S, Lightman SL, Shanks NM. Chronic stress in elderly carers of dementia patients and antibody response to influenza vaccination. Lancet 1999; 353:627-31; PMID:10030328; https://doi.org/10.1016/S0140-6736(98)06098-X
  • Potter JM, O'Donnell B, Carman WF, Roberts MA, Stott DJ. Serological response to influenza vaccination and nutritional and functional status of patients in geriatric medical long-term care. Age Aging 1999; 28:141-5; https://doi.org/10.1093/ageing/28.2.141
  • Falsey AR, Treanor JJ, Tornieporth N, Capellan J, Gorse GJ. Randomized, double-blind controlled phase 3 trial comparing the immunogenicity of high-dose and standard-dose influenza vaccine in adults 65 years of age and older. J Infect Dis 2009; 200:172-80; PMID:19508159; https://doi.org/10.1086/599790
  • 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; 198:650-8; PMID:18652550; https://doi.org/10.1086/590434
  • Podda A. The adjuvanted influenza vaccines with novel adjuvants: experience with the MF59-adjuvanted vaccine. Vaccine 2001; 19:2673-80; PMID:11257408; https://doi.org/10.1016/S0264-410X(00)00499-0
  • Chen WH, Cross AS, Edelman R, Sztein MB, Blackwelder WC, Pasetti MF. Antibody and Th1-type cell-mediated immune responses in elderly and young adults immunized with the standard or a high dose influenza vaccine. Vaccine 2011; 29(16):2865-73; PMID:21352939; https://doi.org/10.1016/j.vaccine.2011.02.017
  • Mannick JB, Del Giudice G, Lattanzi M, Valiante NM, Praestgaard J, Huang B, Lonetto MA, Maecker HT, Kovarik J, Carson S, et al. mTOR inhibition improves immune function in the elderly. Sci Transl Med 2014; 6(268):268ra179; PMID:25540326; https://doi.org/10.1126/scitranslmed.3009892
  • Keating R, Hertz T, Wehenkel M, Harris TL, Edwards BA, McClaren JL, Brown SA, Surman S, Wilson ZS, Bradley P, et al. The kinase mTOR modulates the antibody response to provide cross-protective immunity to lethal infection with influenza virus. Nat Immunol 2013; 14:1266-76; PMID:24141387; https://doi.org/10.1038/ni.2741
  • 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; 82:653-64; PMID:17971814; https://doi.org/10.1038/sj.clpt.6100415
  • 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; 32:1661-9; PMID:24513009; https://doi.org/10.1016/j.vaccine.2014.01.057
  • Linnik JE, Egli A. Impact of host genetic polymorphisms on vaccine induced antibody response. Hum Vaccin Immunother 2016; 12:907-15; PMID:26809773; https://doi.org/10.1080/21645515.2015.1119345
  • 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; 185:114-7; PMID:11756990; https://doi.org/10.1086/338014
  • 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; PMID:23951151; https://doi.org/10.1371/journal.pone.0071376
  • Poland GA, Ovsyannikova IG, Jacobson RM. Immunogenetics of seasonal influenza vaccine response. Vaccine 2008; 26(Suppl 4):D35-40; PMID:19230157; https://doi.org/10.1016/j.vaccine.2008.07.065
  • 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; 10(12):e1004556; PMID:25503988; https://doi.org/10.1371/journal.ppat.1004556
  • Franco LM, Bucasas KL, Wells JM, Niño D, Wang X, Zapata GE, Arden N, Renwick A, Yu P, Quarles JM, et al. Integrative genomic analysis of the human immune response to influenza vaccination. Elife 2013; 2:e00299; PMID:23878721; https://doi.org/10.7554/eLife.00299
  • Klein SL, Jedlicka A, Pekosz A. The Xs and Y of immune responses to viral vaccines. Lancet Infect Dis 2010; 10:338-49; PMID:; PMID:20417416; https://doi.org/10.1016/S1473-3099(10)70049-9
  • Giefing-Kröll C, Berger P, Lepperdinger G, Grubeck-Loebenstein B. How sex and age affect immune responses, susceptibility to infections, and response to vaccination. Aging Cell 2015; 14:309-21; PMID:25720438; https://doi.org/10.1111/acel.12326
  • 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; 168:2405-14; PMID:19064822; https://doi.org/10.1001/archinternmed.2008.513
  • Klein SL, Flanagan KL. Sex differences in immune responses. Nat Rev Immunol 2016; 16:626-38; PMID:27546235; https://doi.org/10.1038/nri.2016.90
  • Furman D, Hejblum BP, Simon N, Jojic V, Dekker CL, Thiébaut R, Tibshirani RJ, Davis MM. Systems analysis of sex differences reveals an immunosuppressive role for testosterone in the response to influenza vaccination. Proc Natl Acad Sci U S A 2014; 111:869-74; PMID:24367114; https://doi.org/10.1073/pnas.1321060111
  • Fish EN. The X-files in immunity: sex-based differences predispose immune responses. Nat Rev Immunol 2008; 8:737-44; PMID:18728636; https://doi.org/10.1038/nri2394
  • Ghorai A, Ghosh U. miRNA gene counts in chromosomes vary widely in a species and biogenesis of miRNA largely depends on transcription or post-transcriptional processing of coding genes. Front Genet 2014; 5:100; PMID:24808907; https://doi.org/10.3389/fgene.2014.00100
  • Morgan OW, Bramley A, Fowlkes A, Freedman DS, Taylor TH, Gargiullo P, Belay B, Jain S, Cox C, Kamimoto L, et al. Morbid obesity as a risk factor for hospitalization and death due to 2009 pandemic influenza A(H1N1) disease. PLoS One 2010; 5(3):e9694; PMID:20300571; https://doi.org/10.1371/journal.pone.0009694
  • Yang L, Chan KP, Lee RS, Chan WM, Lai HK, Thach TQ, Chan KH, Lam TH, Peiris JS, Wong CM. Obesity and influenza associated mortality: evidence from an elderly cohort in Hong Kong. Prev Med 2013; 56:118-23; PMID:23219760; https://doi.org/10.1016/j.ypmed.2012.11.017
  • Azziz-Baumgartner E, Cabrera AM, Chang L, Calli R, Kusznierz G, Baez C, Yedlin P, Zamora AM, Cuezzo R, Sarrouf EB, et al. Mortality, severe acute respiratory infection, and influenza-like ill-ness associated with influenza A(H1N1)pdm09 in Argentina, 2009. PLoS One 2012; 7(10):e47540; https://doi.org/10.1371/journal.pone.0047540
  • 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) 2013; 21:2377-86; PMID:23512822; https://doi.org/10.1002/oby.20383
  • Kim YH, Kim JK, Kim DJ, Nam JH, Shim SM, Choi YK, Lee CH, Poo H. Diet-induced obesity dramatically reduces the efficacy of a 2009 pandemic H1N1 vaccine in a mouse model. J Infect Dis 2012; 205:244-51; https://doi.org/10.1093/infdis/jir731
  • Park HL, Shim SH, Lee EY, Cho W, Park S, Jeon HJ, Ahn SY, Kim H, Nam JH. Obesity-induced chronic inflammation is associated with the reduced efficacy of influenza vaccine. Hum Vaccin Immunother 2014; 10:1181-6; PMID:24614530; https://doi.org/10.4161/hv.28332
  • Karlsson EA, Hertz T, Johnson C, Mehle A, Krammer F, Schultz-Cherry S. Obesity outweighs protection conferred by adjuvanted influenza vaccination. MBio 2016; 7(4):pii: e01144-16; https://doi.org/10.1128/mBio.01144-16
  • 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 (Lond) 2012; 36:1072-7; PMID:22024641; https://doi.org/10.1038/ijo.2011.208
  • Frasca D, Ferracci F, Diaz A, Romero M, Lechner S, Blomberg BB. Obesity decreases B cell responses in young and elderly individuals. Obesity (Silver Spring) 2016; 24:615-25; PMID:26857091; https://doi.org/10.1002/oby.21383
  • Talbot HK, Coleman LA, Crimin K, Zhu Y, Rock MT, Meece J, Shay DK, Belongia EA, Griffin MR. Association between obesity and vulnerability and serologic response to influenza vaccination in older adults. Vaccine 2012; 30:3937-43; PMID:22484350; https://doi.org/10.1016/j.vaccine.2012.03.071
  • Callahan ST, Wolff M, Hill HR, Edwards KM, NIAID Vaccine and Treatment Evaluation Unit (VTEU) Pandemic H1N1 Vaccine Study Group. Impact of body mass index on immunogenicity of pandemic H1N1 vaccine in children and adults. J Infect Dis 2014; 210:1270-4; PMID:24795475; https://doi.org/10.1093/infdis/jiu245
  • Esposito S, Giavoli C, Trombetta C, Bianchini S, Montinaro V, Spada A, Montomoli E, Principi N. Immunogenicity, safety and tolerability of inactivated trivalent influenza vaccine in overweight and obese children. Vaccine 2016; 34:56-60; PMID:26608327; https://doi.org/10.1016/j.vaccine.2015.11.019
  • Abu-Shakra M, Press J, Varsano N, Levy V, Mendelson E, Sukenik S, Buskila D. Specific antibody response after influenza immunization in systemic lupus erythematosus. J Rheumatol 2002; 29:2555-7; PMID:12465151
  • Holvast A, van Assen S, de Haan A, Huckriede A, Benne CA, Westra J, Palache A, Wilschut J, Kallenberg CG, Bijl M. Effect of a second, booster, influenza vaccination on antibody responses in quiescent systemic lupus erythematosus: an open, prospective, controlled study. Rheumatology (Oxford) 2009; 48:1294-9; PMID:19692457; https://doi.org/10.1093/rheumatology/kep200
  • Holvast B, Huckriede A, Kallenberg CG, Bijl M. Influenza vaccination in systemic lupus erythematosus: safe and protective? Autoimmun Rev 2007; 6:300-5
  • Kaur K, Zheng NY, Smith K, Huang M, Li L, Pauli NT, Henry Dunand CJ, Lee JH, Morrissey M, Wu Y, et al. High affinity antibodies against influenza characterize the plasmablast response in SLE patients after vaccination. PLoS One 2015; 10(5):e0125618; PMID:25951191; https://doi.org/10.1371/journal.pone.0125618
  • Del Porto F, Laganà B, Biselli R, Donatelli I, Campitelli L, Nisini R, Cardelli P, Rossi F, D'Amelio R. Influenza vaccine administration in patients with systemic lupus erythematosus and rheumatoid arthritis. Safety and immunogenicity. Vaccine 2006; 24:3217-23; PMID:16466833; https://doi.org/10.1016/j.vaccine.2006.01.028
  • Salemi S, Picchianti-Diamanti A, Germano V, Donatelli I, Di Martino A, Facchini M, Nisini R, Biselli R, Ferlito C, Podestà E, et al. Influenza vaccine administration in rheumatoid arthritis patients under treatment with TNFalpha blockers: safety and immunogenicity. Clin Immunol 2010; 134:113-20; PMID:19846344; https://doi.org/10.1016/j.clim.2009.09.014
  • Oren S, Mandelboim M, Braun-Moscovici Y, Paran D, Ablin J, Litinsky I, Comaneshter D, Levartovsky D, Mendelson E, Azar R, et al. Vaccination against influenza in patients with rheumatoid arthritis: the effect of rituximab on the humoral response. Ann Rheum Dis 2008; 67:937-41; PMID:17981914; https://doi.org/10.1136/ard.2007.077461
  • Lu CC, Wang YC, Lai JH, Lee TS, Lin HT, Chang DM. A/H1N1 influenza vaccination in patients with systemic lupus erythematosus: safety and immunity. Vaccine 2011; 29:444-50; PMID:21078406; https://doi.org/10.1016/j.vaccine.2010.10.081
  • Liao Z, Tang H, Xu X, Liang Y, Xiong Y, Ni J. Immunogenicity and safety of influenza vaccination in systemic lupus erythematosus patients compared with healthy controls: A meta-analysis. PLoS One 2016; 11(2):e0147856; PMID:26845680; https://doi.org/10.1371/journal.pone.0147856
  • Agency EM. Note for guidance on harmonization of requirements for influenza vaccine. London: European Medicine Agency; 1996
  • Pollyea DA, Brown JM, Horning SJ. Utility of influenza vaccination for oncology patients. J Clin Oncol 2010; 28:2481-90; PMID:20385981; https://doi.org/10.1200/JCO.2009.26.6908
  • Ortbals DW, Liebhaber H, Presant CA, Van Amburg AL III, Lee JY. Influenza immunization of adult patients with malignant diseases. Ann Intern Med 1977; 87:552-7; PMID:921082; https://doi.org/10.7326/0003-4819-87-5-552
  • Rubin LG, Levin MJ, Ljungman P, Davies EG, Avery R, Tomblyn M, Bousvaros A, Dhanireddy S, Sung L, Keyserling H, Kang I. 2013 IDSA clinical practice guideline for vaccination of the immunocompromised host. Clin Infect Dis 2014; 58:e44-100; https://doi.org/10.1093/cid/cit684
  • Keam B, Kim MK, Choi Y, Choi SJ, Choe PG, Lee KH, Kim TM, Kim TY, Oh DY, Kim DW, et al. Optimal timing of influenza vaccination during 3-week cytotoxic chemotherapy cycles. Cancer 2016; 123(5):841-8. [ Epub ahead of print]
  • Ceravolo A, Orsi A, Parodi V, Ansaldi F. Influenza vaccination in HIV-positive subjects: latest evidence and future perspective. J Prev Med Hyg 2013; 54:1-10; PMID:24396998
  • Tasker SA, Treanor JJ, Paxton WB, Wallace MR. Efficacy of influenza vaccination in HIV-infected persons. A randomized, double-blind, placebo-controlled trial. Ann Intern Med 1999; 131:430-3; PMID:10498559; https://doi.org/10.7326/0003-4819-131-6-199909210-00006
  • Fuller JD, Craven DE, Steger KA, Cox N, Heeren TC, Chernoff D. Influenza vaccination of human immunodeficiency virus (HIV)-infected adults: impact on plasma levels of HIV type 1 RNA and determinants of antibody response. Clin Infect Dis 1999; 28:541-7; PMID:10194075; https://doi.org/10.1086/515170
  • Kroon FP, van Dissel JT, de Jong JC, Zwinderman K, van Furth R. Antibody response after influenza vaccination in HIV-infected individuals: a consecutive 3-year study. Vaccine 2000; 18:3040-9; PMID:10825608; https://doi.org/10.1016/S0264-410X(00)00079-7
  • Cagigi A, Nilsson A, Pensieroso S, Chiodi F. Dysfunctional B-cell responses during HIV-1 infection: implication for influenza vaccination and highly active antiretroviral therapy. Lancet Infect Dis 2010; 10:499-503; PMID:20610332; https://doi.org/10.1016/S1473-3099(10)70117-1
  • Rinaldi S, Zangari P, Cotugno N, Manno EC, Brolatti N, Castrucci MR, Donatelli I, Rossi P, Palma P, Cagigi A. Antibody but not memory B-cell responses are tuned-down in vertically HIV-1 infected children and young individuals being vaccinated yearly against influenza. Vaccine 2014; 32:657-63; PMID:24333344; https://doi.org/10.1016/j.vaccine.2013.12.008
  • Wheatley AK, Kristensen AB, Lay WN, Kent SJ. HIV-dependent depletion of influenza-specific memory B cells impacts B cell responsiveness to seasonal influenza immunisation. Sci Rep 2016; 6:26478; PMID:27220898; https://doi.org/10.1038/srep26478
  • Rinaldi S, Cagigi A, Santilli V, Zotta F, di Martino A, Castrucci MR, Donatelli I, Poggi E, Piazza A, Campana A, et al. B-sides serologic markers of immunogenicity in kidney transplanted patients: report from 2012–2013 flu vaccination experience. Transplantation 2014; 98:259-66; PMID:24911036; https://doi.org/10.1097/TP.0000000000000209
  • Nath KD, Burel JG, Shankar V, Pritchard AL, Towers M, Looke D, Davies JM, Upham JW. Clinical factors associated with the humoral immune response to influenza vaccination in chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis 2014; 9:51-6; PMID:24399872
  • Valentini D, Marcellini V, Bianchi S, Villani A, Facchini M, atelli I, Castrucci MR, Marasco E, Farroni C, Carsetti R. Generation of switched memory B cells in response to vaccination in Down syndrome children and their siblings. Vaccine 2015; 33:6689-96; PMID:26518399; https://doi.org/10.1016/j.vaccine.2015.10.083
  • Boyd SD, Jackson KJ. Predicting vaccine responsiveness. Cell Host Microbe 2015; 17:301-7; PMID:25766292; https://doi.org/10.1016/j.chom.2015.02.015
  • Hagan T, Nakaya HI, Subramaniam S, Pulendran B. Systems vaccinology: Enabling rational vaccine design with systems biological approaches. Vaccine 2015; 33:5294-301; PMID:25858860; https://doi.org/10.1016/j.vaccine.2015.03.072
  • Killingley B, Enstone JE, Greatorex J, Gilbert AS, Lambkin-Williams R, Cauchemez S, Katz JM, Booy R, Hayward A, Oxford J, et al. Use of a human influenza challenge model to assess person-to-person transmission: proof-of-concept study. J Infect Dis 2012; 205:35-43; PMID:22131338; https://doi.org/10.1093/infdis/jir701
  • Memoli MJ, Czajkowski L, Reed S, Athota R, Bristol T, Proudfoot K, Fargis S, Stein M, Dunfee RL, Shaw PA, et al. Validation of the wild-type influenza A human challenge model H1N1pdMIST: an A(H1N1)pdm09 dose-finding investigational new drug study. Clin Infect Dis 2015; 60(5):693-702; https://doi.org/10.1093/cid/ciu924
  • Krammer F, Palese P. Advances in the development of influenza virus vaccines. Nat Rev Drug Discov 2015; 14:167-82; PMID:25722244; https://doi.org/10.1038/nrd4529
  • de Vries RD, Altenburg AF, Rimmelzwaan GF. Universal influenza vaccines: a realistic option? Clin Microbiol Infect 2016; 22(Suppl 5):S120-S124

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.