Next Article in Journal
Rubella Seroprevalence Boost in the Pediatric and Adolescent Population of Florence (Italy) as a Preventive Strategy for Congenital Rubella Syndrome (CRS)
Previous Article in Journal
Therapeutic Vaccines and Cancer Immunotherapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Exploring the Potential of T-Cells for a Universal Influenza Vaccine

by
Sharon Vijayanand
1,†,
Keegan Braz Gomes
1,†,
Rikhav P. Gala
2,
Mohammad N. Uddin
1 and
Martin J. D’Souza
1,*
1
Vaccine Nanotechnology Laboratory, Mercer University, Atlanta, GA 30341, USA
2
Fraunhofer USA, Center for Molecular Biotechnology, Newark, DE 19711, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Vaccines 2020, 8(4), 598; https://doi.org/10.3390/vaccines8040598
Submission received: 24 September 2020 / Accepted: 30 September 2020 / Published: 11 October 2020
Among the four types of influenza viruses, the influenza A strains and their subtypes have been responsible for causing worldwide pandemics and seasonal epidemics. Around 131 subtypes of the Influenza A virus have been detected and have had their viral genomes mapped [1]. However, creating a vaccine for each specific subtype of the virus is impractical. Moreover, the efficacy of currently existing subtype-specific vaccines, although satisfactory, is affected by several undesirable factors such antigenic drift, mismatch, and pandemic emergence due to assortment. These vaccines are unable to protect most of the population in the event of an emerging novel pandemic strain. Therefore, developing a universal vaccine which would provide cross-protection against the most prominent influenza strains and their subtypes is critical in the near future.
When vaccinated against a specific disease, the adaptive immune system in the body is activated, which is comprised of the T cell and B cell-mediated responses. Therefore, when the actual pathogen attacks the body, it is recognized by the immune system, resulting in a stronger, faster and antigen-specific response, eliminating the invading pathogen. This is the basis of adaptive immunity and T cells play a key role in this process. Recent research has provided promising evidence that T cells evoke a protective as well as long-term immune response [2], capable of mediating heterosubtypic immunity by the recognition of antigenic epitopes conserved across different viral strains and subtypes of the inactivated A virus (IAV) [3]. T cells include the cytotoxic CD8+ T cells and the helper CD4+ T cells, which recognize the antigenic peptides present on the surface of the antigen-presenting cells (APCs) via the major histocompatibility complex (MHC) molecules [4]. In case of a pandemic or seasonal antigenic drift, there may be an insufficient antibody response capable of fighting the infection. In contrast, a T cell vaccine has the potential to produce a memory T cell response, which can recognize a range of dissimilar and even unrelated influenza viruses, by targeting linear peptide sequences conserved across varying influenza virus strains and subtypes [5,6,7,8,9,10]. A preclinical animal study reported that no detectable virus replication was observed when mice were primed with the H3N2 virus, boosted with the H1N1 virus and challenged with the H7N7 virus, demonstrating heterosubtypic protection provided by memory T cells [11]. Accordingly, designing a vaccine that will induce a prominent T cell response, which in turn produces heterosubtypic immunity in an individual, is the basic theory underlying the idea of ‘a universal vaccine’ [3,5].
Several fundamental factors should be taken into consideration prior to developing a universal vaccine, which harnesses the power of T cells. Current vaccine strategies involve the use of surface proteins for vaccine development, which inhibits viral infection in the host but fails to produce conserved T cell epitopes. In contrast, internal proteins are more effectively conserved between different IAV strains and their subtypes, which is of much interest in cross-protection [12,13]. The existing data from human studies report the limited efficacy of both the inactivated influenza vaccine (IIV) and live attenuated influenza vaccine (LAIV), which should be considered while developing a T cell-inducing vaccine [14].
CD4+ helper T cells have also been shown to be critical for establishing both T cell and B cell immune memory. Furthermore, studies in mouse models have shown that CD4+ T cells also provide protection from heterologous influenza infection, which makes CD4+ T cells imperative to future vaccine design [12]. Additionally, the imprinting of hemagglutinin (HA)-subtype-specific CD4+ and B memory cells after prior exposure to various influenza strains can result in the protection from severe infection from similar strains in the future [15]. CD4+ T cells have been shown to offer protection against infection with heterologous influenza strains through their direct cytotoxicity towards influenza infection [16] and reduction in viral loads [17]. Therefore, the effector functions of CD4+ cells including the recalling of heterologous T and B cell responses is highly desirable for developing a universal influenza vaccine.
The role of the T cells in reducing the severity of flu symptoms is supported by ample clinical data. However, standard assay methods to identify a T cell trait that can be attributed to protection is still lacking. T cells act locally, generating a T cell-mediated response when they encounter target cells carrying the antigenic peptide. As the point of entry of the influenza virus is via the respiratory tract, a previously deposited pool of resident memory T cells in the mucosal tissue of the respiratory tract will aid in the prevention of infection and facilitate the elimination of the virus. Tissue resident memory (Trm) in the respiratory tract and lungs respond rapidly to infection by inducing high levels of cytotoxic mediators and effector molecules targeting the virus [18]. An effective vaccine that induces strong influenza virus-specific Trm is a key consideration for developing an effective T cell vaccine. As mentioned previously, internal viral proteins such as the nucleoprotein (NP), polymerase basic 1 protein (PB1), and matrix protein 1 (M1) are highly conserved across different strains of the influenza virus and are therefore capable of producing cross-strain immunity, which is necessary for developing a universal vaccine [5]. Although humans exhibit a diverse expression of MHC alleles and a human leukocyte antigen system (HLA) that varies across ethnicities, T cells generally focus on a select number of peptide + HLA epitopes. However, the search for a universal vaccine should address the current existing ethnic bias in HLA profiles of diverse populations. Variations in T cell responses can be observed depending on the type and class of HLA alleles expressed by the individual [7]. Therefore, studying the HLA profiles of ethnically diverse individuals can contribute to the development of a T cell vaccine that can produce a cross-reactive T cell response, activating multiple immune mechanisms and resulting in an overall robust heterologous immunity.
The influenza virus also contains mechanisms that allow viral escape from the immune-mediated control, which limits the potential of a truly universal influenza vaccine. For example, CD8+ T cell escape has been seen in certain T cell epitopes of naturally circulating influenza virus subtypes [19]. Additionally, preliminary results have confirmed that influenza viral escape can occur very quickly after the virus has infected a mouse [20]. However, evidence suggests that HLA frequencies in a cell population contribute to the rate of T cell-mediated immune escape. To circumvent this issue, various T cell sets can be primed against variants that are likely linked to immune escape [21]. The resulting vaccine strategy would incorporate a modified mix of peptides that could reduce the likelihood of viral escape. However, the specificity of HLA selection for determining the full-length proteins may be unrealistic for large-scale vaccine production. A recent study demonstrated that children infected with human immunodeficiency virus (HIV) were able to generate stronger HIV variant-specific CD8+ T cell responses compared to HIV-infected adults. A similar trend has been reported in children aged 4–14 that have been infected with naturally circulating influenza. Hence, researchers are exploring the significance of combining T cell epitopes, TCR escape variants, and the time of vaccination to produce stronger T cell-inducing influenza vaccines. However, current clinical development does not emphasize the potential of T cell immunity and should therefore be further explored. Certain strategies in clinical development include the use of vectors such as Modified Vaccinia Ankara (MVA) and Simian Adenovirus-encoding internal NP and M1 influenza proteins, which have shown potential to produce robust immunity. The MVA+NP/M1 vaccine has shown positive results in adult and elderly populations and is protective against influenza [22]. T cell-based vaccine strategies have also been evaluated in ferret models, which have shown reduced transmission and lower peak viral loads in vaccinated ferrets [23]. Additionally, viral vectors may possess a self-adjuvanting quality, reducing the need of exogenous adjuvants. Recently, squalene-oil-in-water-based adjuvant MF59 has been approved for use in marketed influenza vaccines and has been reported to improve CD4+ T cell responses post-vaccination [24,25,26]. The use of adjuvants demonstrated strong CD8+ cell responses in ferrets, however, it has not demonstrated the same efficacy in human populations [27].
The review highlighted the underutilized potential of influenza-specific T cell immunity in designing a potential universal vaccine. However, there are some limitations of a T cell approach such as the short life of resident T cells, viral escape, and roles of HLA alleles in population coverage. Also, assessing T cell memory that is influenza-specific in human subjects is more demanding compared to the serology approaches used for assessing antibody responses such as hemagglutination inhibition (HAI) assays. Nonetheless, the potential for T cell recognition against influenza viruses is immense. Antigen development research must also be explored in greater detail to understand its true potential for its application in the development and screening of true universal influenza vaccine candidates that are efficacious and cross-protective against a broad range of influenza strains and their subtypes.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Types of Iinfluenza Viruses, Centers for Disease Control and Prevention, National Center for Immunization and Respiratory Diseases (NCIRD). Available online: https://www.cdc.gov/flu/about/viruses/types.htm (accessed on 20 September 2020).
  2. Bodewes, R.; Kreijtz, J.H.C.M.; Baas, C.; Geelhoed-Mieras, M.M.; De Mutsert, G.; Van Amerongen, G.; Brand, J.M.A.V.D.; Fouchier, R.A.M.; Osterhaus, A.; Rimmelzwaan, G.F. Vaccination against human influenza A/H3N2 virus prevents the induction of heterosubtypic immunity against lethal infection with avian influenza A/H5N1 virus. PLoS ONE 2009, 4, e5538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Grebe, K.M.; Yewdell, J.W.; Bennink, J.R. Heterosubtypic immunity to influenza A virus: Where do we stand? Microbes Infect. 2008, 10, 1024–1029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Croft, N.P.; Purcell, A.W.; Tscharke, D.C. Quantifying epitope presentation using mass spectrometry. Mol. Immunol. 2015, 68, 77–80. [Google Scholar] [CrossRef] [PubMed]
  5. Assarsson, E.; Bui, H.-H.; Sidney, J.; Zhang, Q.; Glenn, J.; Oseroff, C.; Mbawuike, I.N.; Alexander, J.; Newman, M.J.; Grey, H.; et al. Immunomic analysis of the repertoire of T-cell specificities for influenza A virus in humans. J. Virol. 2008, 82, 12241–12251. [Google Scholar] [CrossRef] [Green Version]
  6. Lee, L.Y.-H.; Ha, D.L.A.; Simmons, C.P.; De Jong, M.D.; Chau, N.V.V.; Schumacher, R.; Peng, Y.C.; McMichael, A.J.; Farrar, J.; Smith, G.L.; et al. Memory T cells established by seasonal human influenza A infection cross-react with avian influenza A (H5N1) in healthy individuals. J. Clin. Investig. 2008, 118, 3478–3490. [Google Scholar] [CrossRef]
  7. Quiñones-Parra, S.; Grant, E.; Loh, L.; Nguyen, T.H.O.; Campbell, K.-A.; Tong, S.Y.; Miller, A.; Doherty, P.C.; Vijaykrishna, D.; Rossjohn, J.; et al. Preexisting CD8+ T-cell immunity to the H7N9 influenza A virus varies across ethnicities. Proc. Natl. Acad. Sci. USA 2014, 111, 1049–1054. [Google Scholar] [CrossRef] [Green Version]
  8. Van De Sandt, C.E.; Kreijtz, J.H.C.M.; De Mutsert, G.; Geelhoed-Mieras, M.M.; Hillaire, M.L.B.; Trierum, S.E.V.-V.; Osterhaus, A.D.M.E.; Fouchier, R.A.M.; Rimmelzwaan, G.F. Human cytotoxic T lymphocytes directed to seasonal influenza A viruses cross-react with the newly emerging H7N9 virus. J. Virol. 2013, 88, 1684–1693. [Google Scholar] [CrossRef] [Green Version]
  9. Kreijtz, J.H.C.M.; De Mutsert, G.; Van Baalen, C.A.; Fouchier, R.A.M.; Osterhaus, A.D.M.E.; Rimmelzwaan, G.F. Cross-Recognition of avian H5N1 influenza virus by human cytotoxic T-Lymphocyte populations directed to human influenza A virus. J. Virol. 2008, 82, 5161–5166. [Google Scholar] [CrossRef] [Green Version]
  10. Hillaire, M.L.B.; Trierum, S.E.V.-V.; Kreijtz, J.H.C.M.; De Mutsert, G.; Fouchier, R.A.M.; Osterhaus, A.; Rimmelzwaan, G.F. Human T-cells directed to seasonal influenza A virus cross-react with 2009 pandemic influenza A (H1N1) and swine-origin triple-reassortant H3N2 influenza viruses. J. Gen. Virol. 2013, 94, 583–592. [Google Scholar] [CrossRef] [Green Version]
  11. Christensen, J.P.; Doherty, P.C.; Branum, K.C.; Riberdy, J.M. Profound protection against respiratory challenge with a lethal H7N7 influenza A virus by increasing the magnitude of CD8+ T-cell memory. J. Virol. 2000, 74, 11690–11696. [Google Scholar] [CrossRef] [Green Version]
  12. Valkenburg, S.A.; Li, O.T.W.; Mak, P.W.Y.; Mok, C.K.P.; Nicholls, J.M.; Guan, Y.; Waldmann, T.A.; Peiris, J.S.M.; Perera, L.P.; Poon, L.L.M. IL-15 adjuvanted multivalent vaccinia-based universal influenza vaccine requires CD4+ T cells for heterosubtypic protection. Proc. Natl. Acad. Sci. USA 2014, 111, 5676–5681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Machkovech, H.M.; Bedford, T.; Suchard, M.A.; Bloom, J.D. Positive selection in CD8+ T-cell epitopes of influenza virus nucleoprotein revealed by a comparative analysis of human and swine viral lineages. J. Virol. 2015, 89, 11275–11283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Pebody, R.; McMenamin, J.; Nohynek, H. Live attenuated influenza vaccine (LAIV): Recent effectiveness results from the USA and implications for LAIV programmes elsewhere. Arch. Dis. Child. 2017, 103, 101–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Gostic, K.M.; Ambrose, M.; Worobey, M.; Lloyd-Smith, J.O. Potent protection against H5N1 and H7N9 influenza via childhood hemagglutinin imprinting. Science 2016, 354, 722–726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Brown, D.M.; Dilzer, A.M.; Meents, D.L.; Swain, S.L. CD4 T cell-mediated protection from lethal influenza: Perforin and antibody-mediated mechanisms give a one-two punch. J. Immunol. 2006, 177, 2888–2898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Wilkinson, T.M.; Li, C.K.F.; Chui, C.S.C.; Huang, A.K.Y.; Perkins, M.; Liebner, J.C.; Lambkin-Williams, R.; Gilbert, A.; Oxford, J.; Nicholas, B.; et al. Preexisting influenza-specific CD4+ T cells correlate with disease protection against influenza challenge in humans. Nat. Med. 2012, 18, 274–280. [Google Scholar] [CrossRef]
  18. Mackay, L.K.; Stock, A.T.; Ma, J.Z.; Jones, C.M.; Kent, S.J.; Mueller, S.N.; Heath, W.R.; Carbone, F.R.; Gebhardt, T. Long-lived epithelial immunity by tissue-resident memory T (TRM) cells in the absence of persisting local antigen presentation. Proc. Natl. Acad. Sci. USA 2012, 109, 7037–7042. [Google Scholar] [CrossRef] [Green Version]
  19. Valkenburg, S.A.; Josephs, T.M.; Clemens, E.B.; Grant, E.J.; Nguyen, T.H.O.; Wang, G.C.; Price, A.D.; Miller, A.; Tong, S.Y.; Thomas, P.G.; et al. Molecular basis for universal HLA-A*0201–restricted CD8+ T-cell immunity against influenza viruses. Proc. Natl. Acad. Sci. USA 2016, 113, 4440–4445. [Google Scholar] [CrossRef] [Green Version]
  20. Valkenburg, S.A.; Quiñones-Parra, S.; Gras, S.; Komadina, N.; McVernon, J.; Wang, Z.; Halim, H.; Iannello, P.; Cole, C.; Laurie, K.L.; et al. Acute emergence and reversion of influenza A virus quasispecies within CD8+ T cell antigenic peptides. Nat. Commun. 2013, 4, 1–10. [Google Scholar] [CrossRef] [Green Version]
  21. Valkenburg, S.A.; Gras, S.; Guillonneau, C.; Hatton, L.A.; Bird, N.A.; Twist, K.-A.; Halim, H.; Jackson, D.; Purcell, A.W.; Turner, S.J.; et al. Preemptive priming readily overcomes structure-based mechanisms of virus escape. Proc. Natl. Acad. Sci. USA 2013, 110, 5570–5575. [Google Scholar] [CrossRef] [Green Version]
  22. Berthoud, T.K.; Hamill, M.; Lillie, P.J.; Hwenda, L.; Collins, K.A.; Ewer, K.J.; Milicic, A.; Poyntz, H.C.; Lambe, T.; Fletcher, H.A.; et al. Potent CD8+ T-cell immunogenicity in humans of a novel heterosubtypic influenza A vaccine, MVA-NP+M1. Clin. Infect. Dis. 2011, 52, 1–7. [Google Scholar] [CrossRef] [PubMed]
  23. Baz, M.; Boonnak, K.; Paskel, M.; Santos, C.; Powell, T.J.; Townsend, A.; Subbarao, K. Nonreplicating Influenza A Virus Vaccines Confer Broad Protection against Lethal Challenge. mBio 2015, 6, e01487-15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Giarola-Silva, S.; Coelho-Dos-Reis, J.G.; Mourão, M.M.; Campi-Azevedo, A.C.; Silva, E.E.N.; Luiza-Silva, M.; Martins, M.A.; Silveira-Cassette, A.C.D.O.; Batista, M.A.; Peruhype-Magalhães, V.; et al. Distinct patterns of cellular immune response elicited by influenza non-adjuvanted and AS03-adjuvanted monovalent H1N1(pdm09) vaccine. Antivir. Res. 2017, 144, 70–82. [Google Scholar] [CrossRef]
  25. O’Hagan, D.; Rappuoli, R.; De Gregorio, E.; Tsai, T.F.; Del Giudice, G. MF59 adjuvant: The best insurance against influenza strain diversity. Expert Rev. Vaccines 2011, 10, 447–462. [Google Scholar] [CrossRef] [PubMed]
  26. Couch, R.B.; Rodríguez, J.M.B.; Caso, C.; Mbawuike, I.N.; López, C.N.; Claeys, C.; El Idrissi, M.; Hervé, C.; Laupeze, B.; Oostvogels, L.; et al. Superior antigen-specific CD4+ T-cell response with AS03-adjuvantation of a trivalent influenza vaccine in a randomised trial of adults aged 65 and older. BMC Infect. Dis. 2014, 14, 425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Kumar, A.; McElhaney, J.E.; Walrond, L.; Cyr, T.D.; Merani, S.; Kollmann, T.R.; Halperin, S.A.; Scheifele, D.W. Cellular immune responses of older adults to four influenza vaccines: Results of a randomized, controlled comparison. Hum. Vaccines Immunother. 2017, 13, 2048–2057. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Vijayanand, S.; Gomes, K.B.; Gala, R.P.; Uddin, M.N.; D’Souza, M.J. Exploring the Potential of T-Cells for a Universal Influenza Vaccine. Vaccines 2020, 8, 598. https://doi.org/10.3390/vaccines8040598

AMA Style

Vijayanand S, Gomes KB, Gala RP, Uddin MN, D’Souza MJ. Exploring the Potential of T-Cells for a Universal Influenza Vaccine. Vaccines. 2020; 8(4):598. https://doi.org/10.3390/vaccines8040598

Chicago/Turabian Style

Vijayanand, Sharon, Keegan Braz Gomes, Rikhav P. Gala, Mohammad N. Uddin, and Martin J. D’Souza. 2020. "Exploring the Potential of T-Cells for a Universal Influenza Vaccine" Vaccines 8, no. 4: 598. https://doi.org/10.3390/vaccines8040598

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop