Elsevier

Vaccine

Volume 26, Issue 16, 7 April 2008, Pages 1990-1998
Vaccine

Discordance between antibody and T cell responses in recipients of trivalent inactivated influenza vaccine

https://doi.org/10.1016/j.vaccine.2008.02.024Get rights and content

Summary

Thirty adults were tested for humoral and cellular immune responses following immunization with the trivalent inactivated influenza vaccine. Modest but significant inverse correlations between the baseline and the fold changes in the number of IFNγ-producing cells and the levels of neutralizing antibodies were observed. Specific increases in proliferative responses in the CD8 CD45RA+ population were noted after vaccination. Minimal correlations between neutralizing antibody titers and the number of IFNγ-producing cells in terms of prevaccination levels or fold increases were observed. These results show specific increases in a CD8 T cell subset and discordant T and B responses induced by the trivalent inactivated influenza vaccine.

Introduction

Human influenza is a highly contagious acute respiratory illness that is responsible for significant morbidity and excess mortality in the elderly and the very young worldwide. Though effective antiviral medications targeting the neuraminidase (NA) glycoprotein are available, prevention of influenza morbidity and mortality is primarily through the immunization of target groups at high risk for mortality or hospitalization [1]. Annual worldwide epidemics of influenza A and the recent emergence of zoonotic infections with highly pathogenic H5N1 and H9N2 avian influenza strains have heightened efforts to understand the role of both humoral and cell-mediated immunity in the control of influenza virus infection [2].

Current vaccine approaches depend on the induction of antibodies to the viral surface proteins hemagglutinin and neuraminidase that neutralize the infectivity of the virus and interfere with the release of newly replicated virus from the host cell [2], [3]. When the vaccine virus closely matches the challenge infecting virus, the vaccines are effective. The virus however undergoes frequent mutations at antibody combining sites and the vaccine is less effective. This is a much bigger problem when a new subtype of influenza virus emerges, e.g. the H5N1 avian derived viruses, and there are no cross-reactive antibody sites on the vaccine virus hemagglutinin (HA) and the new subtype of virus. Ideally, influenza vaccines would also be expected to induce influenza specific CD8 T cell-mediated responses that may contribute to protective immunity. Studies in murine models of influenza demonstrated that CD8 T cells were effective in reducing viral titers and aided in recovery [4], [5], [6], [7], [8], [9]. These models have also demonstrated that delayed influenza virus clearance occurs in CD8 T cell deficient mice [10] and that memory T lymphocytes can act independently of a humoral immune response in order to confer resistance to influenza infection [11], [12]. Influenza virus-specific cytotoxic T lymphocytes (CTL) have also been shown to limit influenza A virus replication and protect against lethal viral challenge [7], [8], [13]. The role of CD4 T cells in influenza infection is less defined. CD4 virus-specific T cells may help compensate for the absence of CD8 CTL because the virus can be cleared in CD8-deficient mice; however, mice lacking both CD4 and CD8 T cells do not clear virus or survive [14], [15]. Belz et al. demonstrated that responses in the secondary lymphoid organs of CD4 T cell deficient mice infected with influenza were defective while responses in bronchoalveolar lavage were similar in CD4 T cell deficient mice and wild type mice. This suggested that CD4 T cells may not be required for the primary response to influenza but may be important in the generation of memory CD8 T cells [16].

In contrast, there have been few studies on cellular immunity to influenza in humans. Knowledge about human CD8 T cell immune responses has been more well developed than human CD4 T cell responses [17], [18]. Studies on the cytotoxic T lymphocyte repertoire to influenza A viruses indicate that influenza memory T cell responses are directed to a number of epitopes on a variety of proteins including the nucleoprotein, nonstructural protein 1 (NS1), and the matrix protein 1 (M1) [19], [20]. Most of these highly conserved cross-reactive epitopes have been found to be conserved in H5N1 viruses from recent outbreaks [17]. Therefore, cell-mediated immunity appears to be important in both restricting influenza A virus replication and reducing disease severity, and appears to offer a more potentially cross-reactive vaccine approach for the prevention of pandemic or epidemic influenza.

In this study, we evaluated the human memory T cell and the serum antibody responses of healthy subjects following immunization with the licensed 2005–2006 trivalent inactivated influenza vaccine in order to better understand the role that both T and B cells may play in the protection afforded by the vaccine. Using carboxyfluoroscein succinimidyl ester (CFSE), enzyme-linked immunosorbent spot (ELISPOT) and neutralization assays to examine responses to the individual influenza A H1 and H3 viral strains, we were particularly interested in the following questions: (1) the effect of prevaccination levels of T cell immunity to both influenza A virus strains on the cellular immune responses generated by subsequent vaccination, (2) the phenotype(s) of the T cells responding to vaccine and (3) the relationships between the antibody and T cell components of the host immune response to influenza vaccine.

Section snippets

Viruses

Influenza A viruses H1N1 A/New Caledonia/20/99 (Lot # 3XANA060818B) and H3N2 A/Wisconsin/67/05 (Lot # 3XAWN060818B) were purchased from Charles River Laboratories (North Franklin, Connecticut) for use in CFSE, ELISPOT, and proliferation assays. These viruses were propagated in the allantoic cavity in Specific Pathogen Free (SPF) eggs with the final hemagglutinin titers for the A/New Caledonia strain and the A/Wisconsin strains 1:512 per 0.05 ml and 1:16 per 0.05 ml, respectively. The following

IFNγ-producing cell responses to influenza vaccination

We used ELISPOT assays to quantitate the number of IFNγ-producing cells in PBMC specific for the influenza A H1N1 subtype strain A/New Caledonia/20/99 and the H3N2 subtype strain A/Wisconsin/67/05. The H3N2 A/Wisconsin/67/05 virus is a very closely related antigenic variant of the 2005–06 vaccine strain H3N2A/California/07/04. There was an overall moderate but not a statistically significant increase in the number of IFN-γ-producing cells post-vaccination after stimulation with either of the

Discussion

Our study examined the pattern of cellular responses in influenza vaccinated individuals using three different T cell assays, ELISPOT to quantitate the numbers of specific IFNγ-producing cells and 3H thymidine and CFSE assays to quantitate the number of proliferating T cells, in relation to B cell responses examined using microneutralization assays. We studied the responses to two influenza A subtype viruses—H1N1 A/New Caledonia/20/99 and H3N2 A/Wisconsin/67/05 which contain the HA and NA

Acknowledgements

We thank Dr. Jeffrey S. Kennedy, Karen Longtine, Melissa O’Neill, and Jaclyn Longtine for their help in obtaining the human PBMC samples that were used in this study. We thank Christine Turcotte for assistance with HLA typing and Ping Liu for preliminary experiments using plaque assays as a readout for the neutralization assay.

References (40)

  • Y.K. Yamada et al.

    Human interferon alpha and gamma production by lymphocytes during the generation of influenza virus-specific cytotoxic T lymphocytes

    J Gen Virol

    (1986)
  • B.A. Askonas et al.

    An influenza specific T-killer clone is restricted to H-2Ld and cross-reacts with Dk region

    Immunogenetics

    (1982)
  • K. Kuwano et al.

    HA2 subunit of influenza A H1 and H2 subtype viruses induces a protective cross-reactive cytotoxic T lymphocyte response

    J Immunol

    (1988)
  • K. Kuwano et al.

    Cytotoxic T lymphocytes recognize a cross-reactive epitope on the transmembrane region of influenza H1 and H2 hemagglutinins

    Viral Immunol

    (1989)
  • B.S. Bender et al.

    Transgenic mice lacking class I major histocompatibility complex-restricted T cells have delayed viral clearance and increased mortality after influenza virus challenge

    J Exp Med

    (1992)
  • M.B. Graham et al.

    Resistance to and recovery from lethal influenza virus infection in B lymphocyte-deficient mice

    J Exp Med

    (1997)
  • S.L. Epstein et al.

    Mechanism of protective immunity against influenza virus infection in mice without antibodies

    J Immunol

    (1998)
  • C.D. Mackenzie et al.

    Rapid recovery of lung histology correlates with clearance of influenza virus by specific CD8+ cytotoxic T cells

    Immunology

    (1989)
  • M.A. Wells et al.

    Recovery from a viral respiratory infection. I. Influenza pneumonia in normal and T-deficient mice

    J Immunol

    (1981)
  • M. Eichelberger et al.

    Clearance of influenza virus respiratory infection in mice lacking class I major histocompatibility complex-restricted CD8+ T cells

    J Exp Med

    (1991)
  • Cited by (29)

    • Comparison of neutralizing antibody and cell-mediated immune responses to pandemic H1N1 2009 influenza virus before and after H1N1 2009 influenza vaccination of elderly subjects and healthcare workers

      2012, International Journal of Infectious Diseases
      Citation Excerpt :

      Our study revealed significant increases in T-cell responses after vaccination. However, there was no concordance between the antibody and T-cell responses, a result that concurs with a previous finding in individuals vaccinated with trivalent inactivated influenza vaccine in 2008, suggesting that vaccination does not trigger equivalent humoral and cellular responses.20,22 Moreover, there were important age-related differences in the T-cell responses compared to the antibody responses to vaccination, with older individuals showing both higher pre-vaccination antibody titers and T-cell responses to pH1N1 2009.

    • New technologies for new influenza vaccines

      2012, Vaccine
      Citation Excerpt :

      The literature is replete with studies measuring antibody fold-rise over background and finding that the lower the background, the higher the fold-rise. The same thing applies to T-cell responses [63]. Vaccines intended to induce a T-cell response fall into two general categories; vaccines comprising selected conserved peptides, often as concatamers, and vectored delivery of antigens.

    • Ki-67 expression reveals strong, transient influenza specific CD4 T cell responses after adult vaccination

      2012, Vaccine
      Citation Excerpt :

      Thus influenza TIV vaccination selectively induced Ki-67 expression in the influenza-specific but not bulk CD4 memory T cell populations. Although previous reports [6,7] and our current data suggest that adult TIV vaccination does not significantly alter the overall number of influenza-reactive CD4 memory T cells, we have now shown a striking increase in Ki-67+ cells during the response, in some subjects reaching more than 30% of the cytokine-expressing cells at 4–6 days after vaccination. These apparently paradoxical results can be reconciled if it is assumed that the vaccine actually induces a vigorous memory cell proliferative response, but that this response is balanced by a rapid contraction phase, resulting in little change in overall numbers.

    • Transfer of influenza vaccine-primed costimulated autologous T cells after stem cell transplantation for multiple myeloma leads to reconstitution of influenza immunity: Results of a randomized clinical trial

      2011, Blood
      Citation Excerpt :

      The patients in the primed group trended toward a higher CD8 T-cell response than the patients in the nonprimed group; however, the difference did not reach statistical significance. The inactivated influenza vaccine stimulates predominantly CD4 T-cell responses.45 We, therefore, measured T-cell ELISPOT results after depletion of CD8 T cells and found that CD4-specific responses were significantly better in the primed group at the T-cell harvest time point (P = .02) (Figure 5).

    View all citing articles on Scopus

    Financial support: This work was supported by Funding from the National Institute of Allergy and Infectious Diseases/National Institutes of Health Grant U19 AI-057319.

    View full text