Elsevier

Vaccine

Volume 29, Issue 47, 3 November 2011, Pages 8606-8614
Vaccine

Age-related changes in durability and function of vaccine-elicited influenza-specific CD4+ T-cell responses

https://doi.org/10.1016/j.vaccine.2011.09.019Get rights and content

Abstract

The major antigenic component of licensed influenza vaccines, hemagglutinin (HA), elicits predominantly type-specific antibody responses, thus necessitating frequent antigenic updates to the annual vaccine. However, accumulating evidence suggests that influenza vaccines can also induce significant cross-reactive T-cell responses to highly divergent, heterosubtypic HA antigens not included in the vaccine. Influenza vaccines are less effective among the elderly and studies that characterize cross-reactive T-cell immunity in this vulnerable population are much needed. Here, we systematically compare the ex vivo frequency, cytokine profile and phenotype of vaccine-elicited HA-specific T-cell responses among a cohort of young (18–49 years old) and elderly (≥70 years old) vaccinees, as well as the maturation and activation phenotype of total CD4+ and CD8+ T-cells. IFN-γ production after in vitro expansion and HA-specific Ab titers were also determined. We find that vaccine-elicited ex vivo frequencies of CD4+ T-cells elicited by vaccination reactive to any given homo- or heterosubtypic Ag were comparable across the two age groups. While, no differences were observed between age groups in the phenotype of Ag-specific or total CD4+ T-cells, PBMC from young adults were superior at producing IFN-γ after short-term Ag-specific culture. Significantly, while vaccine-elicited T-cell responses were durable among the younger vaccinees, they were short-lived among the elderly. These results have important ramifications for our understanding of vaccine-induced changes in the magnitude and functionality of HA-specific CD4+ T-cells, as well as age-related alterations in response kinetics.

Highlights

► Influenza vaccine (TIV) immunogenicity among young (18–49 years old) and elderly (≥70 years old). ► Vaccine elicits predominantly central memory and effector memory CD4+ T-cells. ► Vaccine-elicited T cell responses are short-lived among the elderly. ► PBMC from younger subjects are superior at producing IFN-γ after short-term Ag-specific culture.

Introduction

The influenza pandemic of 1918 saw the introduction of an avian-origin H1N1 that is estimated to have caused over 40 million deaths worldwide [1]. The influenza pandemic of 2009 similarly saw the introduction of an antigenically novel H1N1 into the global population and is estimated to have caused over 18,449 deaths globally [2]. In spite of the dominant emergence of this novel swine-origin H1N1 lineage in 2009, multiple instances of lethal avian influenza infections among humans have also occurred over the past decade. While divergent influenza subtypes including H7N7, H9N2, and H7N3 have circulated sporadically, recurrent outbreaks of the highly pathogenic H5N1 have been documented consistently [3], [4], [5]. Though these outbreaks have primarily occurred in south-east Asia, avian influenza infections among humans and birds have also been reported from Central and Western European nations thus increasing the likelihood of a potential H5N1 pandemic emerging in the future [6].

A number of studies in humans [7], [8], [9], [10] as well as in animal model systems [11], [12] suggest that protective immunity against influenza correlates with the prevalence of influenza-specific antibodies, primarily those directed against the viral hemagglutinin (HA). However, humoral immunity to influenza is predominantly type-specific and affords limited protection against heterosubtypic strains of circulating viruses [13]. Thus, the continual drift- and periodic antigenic shift-events that introduce antigenically novel influenza viruses into the global population have required annual changes in influenza vaccines to achieve reliable vaccine-induced protection [14]. However, instances of mismatches between seasonal influenza Ags found in the vaccine and circulating influenza viruses have generally lead to limited vaccine efficacy in some years [13], [15], [16], [17], [18], [19]. Equally, with subtypes such as H5N1 that show future pandemic potential, several distinct heterotypic strains have been involved in the outbreaks to date, thus increasing uncertainty as to which strain or strains may emerge as a potential avian pandemic strain.

With such antigenic plasticity as a backdrop, cellular immune responses are being increasingly recognized as an alternate correlate of immune protection that could provide protection from influenza viruses of disparate antigenic lineages [20]. A number of studies have recently characterized the existence of subtype-independent, cross-reactive cellular immune responses against diverse influenza strains [21], [22], [23], [24], which may provide protection from influenza infection or reduce severity of influenza disease [24], [25]. Importantly, recent studies clearly suggest that among the elderly, a population that bears the brunt of influenza morbidity and mortality, cell mediated immunity provides a better correlate of protection than do serum antibody responses [20]. As with antibody responses, HA is a major component of protective cellular immunity against influenza [26], [27], including heterosubtypic immunity [28].

Accumulating evidence suggests that cross-reactive T-cell responses can be induced de novo or boosted over pre-existing levels following seasonal vaccination with an unmatched HA antigen [23]. Though our earlier study utilizing a cohort of 18–49-year-olds was unable to assess cross-reactive T-cell responses among the truly elderly [29], we found that ageing reduced the magnitude of T-cells cross-reactive to pandemic H1 even in this relatively young cohort. Also, our use of IFN-γ ELISPOT following expansion in vitro precluded the assessment of HA-specific T-cell frequencies, as well as their memory and activation status directly ex vivo. In the present study, polychromatic flow cytometry was used to compare the ex vivo frequency, maturation and activation phenotypes of HA-specific CD4+ T-cells elicited following seasonal influenza vaccine administration among young (18–49 years of age) and elderly (≥70 years of age) adults. As our trial was conducted in 2007, we also address the ability of seasonal vaccination to induce cross-reactive T-cells against antigens that vaccinees had not yet been exposed to, such as the avian H5 that has only seen sporadic outbreaks in Asia, and the novel pandemic swine-origin H1 of 2009 that emerged two seasons following sample collection.

Section snippets

Human subjects and sample collection

This study was performed at Cincinnati Children's Hospital Medical Center in the fall of 2007 before seasonal influenza circulation in the community and two seasons prior to the emergence of the novel pandemic 2009 H1N1 strain globally. A total of 30 subjects were recruited into the study and provided informed consent following appropriate Institutional Review Board (IRB) and Institutional Biosafety Committee evaluation, and included 15 subjects each in the “young” (18–49 years old) and elderly

Age related changes in global CD4+ T-cell differentiation and activation status

While deficits in both magnitude and functionality of influenza-specific T-cells have previously been documented in relation to ageing of the host [32], [33], [34], the mechanisms underlying these deficits are currently not well understood. It is conceivable that this is a result of global changes in the maturation and activation status of T-cell subsets among the elderly that is not specific to influenza. To address this, total T-cell populations of young and elderly vaccinees were assessed by

Discussion

While the preponderance of influenza specific humoral responses is type-specific, a number of studies have now established the existence of robust cross-reactive T-cell responses against heterosubtypic antigens of influenza [21], [22], [23], [24]. We have previously shown that such heterosubtypic T-cell responses can be boosted by vaccination with unmatched antigens among human vaccinees [23]. Importantly, the major antigenic component of all licensed influenza vaccines, HA, has been identified

Acknowledgements

The authors would like to thank Dr. Alex Klimov and Dr. Xiyan Xu at the Center for Disease Control and Prevention (CDC), as well as BEI Resources for the influenza reagents. We thank Dr. David Bernstein, Tarek Shata and Saleem Basha for discussions; Tara Foltz for clinical study coordination; and Joanne Yu for preparation of Ab-conjugates. We also thank Dr. Martha Nason for help with statistical analysis and power calculations. This work was supported by the Intramural Research Program of the

References (62)

  • J.J. Goronzy et al.

    Aging and T-cell diversity

    Exp Gerontol

    (2007)
  • D.M. Murasko et al.

    Role of humoral and cell-mediated immunity in protection from influenza disease after immunization of healthy elderly

    Exp Gerontol

    (2002)
  • P. Gogolak et al.

    Mapping of a protective helper T cell epitope of human influenza A virus hemagglutinin

    Biochem Biophys Res Commun

    (2000)
  • R.B. Effros

    Loss of CD28 expression on T lymphocytes: a marker of replicative senescence

    Dev Comp Immunol

    (1997)
  • I. Saba et al.

    IL-7R-dependent survival and differentiation of early T-lineage progenitors is regulated by the BTB/POZ domain transcription factor Miz-1

    Blood

    (2011)
  • R. Channappanavar et al.

    Advancing age leads to predominance of inhibitory receptor expressing CD4 T cells

    Mech Ageing Dev

    (2009)
  • J.M. Brenchley et al.

    Expression of CD57 defines replicative senescence and antigen-induced apoptotic death of CD8+ T cells

    Blood

    (2003)
  • S.A. Richard et al.

    Evaluation of Southern hemisphere influenza vaccine recommendations

    Vaccine

    (2010)
  • M.C. Steinhoff

    Influenza in young children: burden, immunisation, and policy

    Lancet Infect Dis

    (2011)
  • P. Palese

    Influenza: old and new threats

    Nat Med

    (2004)
  • WHO

    Global Alert and Response (GAR): pandemic (H1N1) 2009 – update 112

    (2010)
  • R.G. Webster et al.

    Microbial adaptation and change: avian influenza

    Rev Sci Tech

    (2004)
  • K. Subbarao et al.

    Avian influenza viruses infecting humans

    Cell Mol Life Sci

    (2000)
  • D. Normile

    Infectious diseases. Genetic analyses suggest bird flu virus is evolving

    Science

    (2005)
  • R.B. Couch et al.

    Immunity to influenza in man

    Annu Rev Microbiol

    (1983)
  • A. Delem

    Serum SRH antibody level as a measure of the immunity against natural and artificial A/Victoria/3/75 infections

    Dev Biol Stand

    (1977)
  • A. Goodeve et al.

    A graded-dose study of inactivated, surface antigen influenza B vaccine in volunteers: reactogenicity, antibody response and protection to challenge virus infection

    J Hyg (Lond)

    (1983)
  • J.R. Davies et al.

    Natural or vaccine-induced antibody as a predictor of immunity in the face of natural challenge with influenza viruses

    Epidemiol Infect

    (1989)
  • J.L. Virelizier

    Host defenses against influenza virus: the role of anti-hemagglutinin antibody

    J Immunol

    (1975)
  • W. Gerhard et al.

    Role of the B-cell response in recovery of mice from primary influenza virus infection

    Immunol Rev

    (1997)
  • J.C. de Jong et al.

    Mismatch between the 1997/1998 influenza vaccine and the major epidemic A(H3N2) virus strain as the cause of an inadequate vaccine-induced antibody response to this strain in the elderly

    J Med Virol

    (2000)
  • Cited by (16)

    View all citing articles on Scopus
    View full text