The human potential of a recombinant pandemic influenza vaccine produced in tobacco plants

Åsne Jul-Larsen, Abdullah S Madhun, Karl A Brokstad, Emanuele Montomoli, Vidadi Yusibov, Rebecca J Cox, Åsne Jul-Larsen, Abdullah S Madhun, Karl A Brokstad, Emanuele Montomoli, Vidadi Yusibov, Rebecca J Cox

Abstract

Rapid production of influenza vaccine antigen is an important challenge when a new pandemic occurs. Production of recombinant antigens in plants is a quick, cost effective and up scalable new strategy for influenza vaccine production. In this study, we have characterized a recombinant influenza haemagglutinin antigen (HAC1) that was derived from the 2009 pandemic H1N1 (pdmH1N1) virus and expressed in tobacco plants. Volunteers vaccinated with the 2009 pdmH1N1 oil-in-water adjuvanted vaccine provided serum and lymphocyte samples that were used to study the immunogenic properties of the HAC1 antigen in vitro. By 7 d post vaccination, the vaccine fulfilled the licensing criteria for antibody responses to the HA detected by haemagglutination inhibition and single radial hemolysis. By ELISA and ELISPOT analysis we showed that HAC1 was recognized by specific serum antibodies and antibody secreting cells, respectively. We conducted a kinetic analysis and found a peak of serum HAC1 specific antibody response between day 14 and 21 post vaccination by ELISA. We also detected elevated production of IL-2 and IFNγ and low frequencies of CD4(+) T cells producing single or multiple Th1 cytokines after stimulating PBMCs (peripheral blood mononuclear cells) with the HAC1 antigen in vitro. This indicates that the antigen can interact with T cells, although confirming an effective adjuvant would be required to improve the T-cell stimulation of plant based vaccines. We conclude that the tobacco derived recombinant HAC1 antigen is a promising vaccine candidate recognized by both B- and T cells.

Figures

https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3495720/bin/hvi-8-653-g1.jpg
Figure 1. Solicited adverse events after vaccination in the enrolled volunteers. The frequency of local and systemic adverse events for volunteers included in the study. Data are based on diary cards completed by the 23 subjects in the study. “Any” refers to the report of an adverse event on one or more days after vaccination.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3495720/bin/hvi-8-653-g2.jpg
Figure 2. Immunogenicity of the pdmH1N1 vaccine. (A) serum SRH titers for the enrolled volunteers pre-vaccination, and 7, 14, 21 d (d) post vaccination. (B) serum HI titers for the enrolled volunteers pre-vaccination, and 7, 14, 21 d (d) and 3, 6 and 12 mo (m) post vaccination. Each symbol in A and B represents one individual serum sample and lines indicate geometric mean area (GMA) or geometric mean titer (GMT) ± 95% confidence interval. The number of sera (n) included at each time point is indicated. (C) The reverse cumulative distribution curves for the HI antibody response. The HI data was standardized according to a conversion factor based on the GMT of the Candidate International Standard (09/194) in a collaborative study, as the Candidate International Standard has not been assigned International Units. Dotted lines indicate protective levels as defined by the Committee for Medicinal Products for Human Use (CHMP).
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3495720/bin/hvi-8-653-g3.jpg
Figure 3. Human B-cells detecting HAC1. Human peripheral blood mononuclear cells (PBMCs) isolated 7 d post vaccination were analyzed by ELISPOT to identify antibody secreting cells (ASCs) recognizing the HAC1 antigen or the vaccine antigen. For each sample the number of B-cells secreting specific antibodies of the IgG, IgA and IgM classes were quantified in 100,000 PBMCs. Each symbol represents one individual volunteer. Lines indicate the mean ± standard error of the mean (SEM). n = 15
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3495720/bin/hvi-8-653-g4.jpg
Figure 4. Human serum antibodies detecting HAC1. HAC1 detecting IgG antibodies were measured by ELISA on serum collected pre vaccination and at 7, 14 and 21 d (d) and 3, 6 and 12 mo (m) post vaccination. Each symbol represents one individual serum sample. Lines represent the mean ± standard error of the mean (SEM). ** and *** indicate concentrations significantly different from day 0 with p < 0.005 and p < 0.0005, respectively. The number of sera (n) included at each time point is indicated.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3495720/bin/hvi-8-653-g5.jpg
Figure 5. In vitro induction of cytokine secretion from human PBMCs by HAC1. Human PBMCs isolated 21 d post vaccination were stimulated in vitro with HAC1 or the vaccine antigen or left non-stimulated (Neg) for 3 d and the level of 10 different cytokines was measured in the cell supernatant by multiplex cytokine assay. Each symbol represents one individual volunteer. Bars represent the mean ± standard error of the mean (SEM). * and ** indicate significant difference with p < 0.05 and p < 0.005, respectively. n = 19.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3495720/bin/hvi-8-653-g6.jpg
Figure 6. HAC1 activation of multifunctional T cells. Human PBMCs isolated 21 d post vaccination were stimulated in vitro with HAC1 or the vaccine antigen or left non-stimulated (Neg) overnight. Cells were then fixed and analyzed for the presence of multifunctional T cells producing one or more of the cytokines TNFα, IL-2 and IFNγ by multi-parametric flow cytometry. (A) Total frequency of CD4+ cells producing each of the cytokines investigated. (B) Frequency of CD4+ cells producing only one, any two or all three of the cytokines investigated. *, ** and *** indicate significant difference with p < 0.05, p < 0.005 and p < 0.0005, respectively. Bars represent the mean ± standard error of the mean (SEM). n = 19.

References

    1. Chichester JA, Yusibov V. Plants as alternative systems for production of vaccines. Hum Vaccin. 2007;3:146–8. doi: 10.4161/hv.3.4.4148.
    1. Chichester JA, Haaheim LR, Yusibov V. Using plant cells as influenza vaccine substrates. Expert Rev Vaccines. 2009;8:493–8. doi: 10.1586/erv.09.3.
    1. Shoji Y, Chichester JA, Jones M, Manceva SD, Damon E, Mett V, et al. Plant-based rapid production of recombinant subunit hemagglutinin vaccines targeting H1N1 and H5N1 influenza. Hum Vaccin. 2011;7(Suppl):41–50. doi: 10.4161/hv.7.0.14561.
    1. Lu X, Tumpey TM, Morken T, Zaki SR, Cox NJ, Katz JM. A mouse model for the evaluation of pathogenesis and immunity to influenza A (H5N1) viruses isolated from humans. J Virol. 1999;73:5903–11.
    1. Lipatov AS, Hoffmann E, Salomon R, Yen H-L, Webster RG. Cross-protectiveness and immunogenicity of influenza A/Duck/Singapore/3/97(H5) vaccines against infection with A/Vietnam/1203/04(H5N1) virus in ferrets. J Infect Dis. 2006;194:1040–3. doi: 10.1086/507709.
    1. Treanor JJ, Wilkinson BE, Masseoud F, Hu-Primmer J, Battaglia R, O’Brien D, et al. Safety and immunogenicity of a recombinant hemagglutinin vaccine for H5 influenza in humans. Vaccine. 2001;19:1732–7. doi: 10.1016/S0264-410X(00)00395-9.
    1. Nicholson KG, Colegate AE, Podda A, Stephenson I, Wood J, Ypma E, et al. Safety and antigenicity of non-adjuvanted and MF59-adjuvanted influenza A/Duck/Singapore/97 (H5N3) vaccine: a randomised trial of two potential vaccines against H5N1 influenza. Lancet. 2001;357:1937–43. doi: 10.1016/S0140-6736(00)05066-2.
    1. Zhu F-C, Wang H, Fang H-H, Yang JG, Lin XJ, Liang X-F, et al. A novel influenza A (H1N1) vaccine in various age groups. N Engl J Med. 2009;361:2414–23. doi: 10.1056/NEJMoa0908535.
    1. Liang X-F, Wang H-Q, Wang J-Z, Fang H-H, Wu J, Zhu F-C, et al. Safety and immunogenicity of 2009 pandemic influenza A H1N1 vaccines in China: a multicentre, double-blind, randomised, placebo-controlled trial. Lancet. 2010;375:56–66. doi: 10.1016/S0140-6736(09)62003-1.
    1. Greenberg ME, Lai MH, Hartel GF, Wichems CH, Gittleson C, Bennet J, et al. Response to a monovalent 2009 influenza A (H1N1) vaccine. N Engl J Med. 2009;361:2405–13. doi: 10.1056/NEJMoa0907413.
    1. Plennevaux E, Sheldon E, Blatter M, Reeves-Hoché M-K, Denis M. Immune response after a single vaccination against 2009 influenza A H1N1 in USA: a preliminary report of two randomised controlled phase 2 trials. Lancet. 2010;375:41–8. doi: 10.1016/S0140-6736(09)62026-2.
    1. Clark TW, Pareek M, Hoschler K, Dillon H, Nicholson KG, Groth N, et al. Trial of 2009 influenza A (H1N1) monovalent MF59-adjuvanted vaccine. N Engl J Med. 2009;361:2424–35. doi: 10.1056/NEJMoa0907650.
    1. Madhun AS, Akselsen PE, Sjursen H, Pedersen G, Svindland S, Nøstbakken JK, et al. An adjuvanted pandemic influenza H1N1 vaccine provides early and long term protection in health care workers. Vaccine. 2010;29:266–73. doi: 10.1016/j.vaccine.2010.10.038.
    1. Shoji Y, Chichester JA, Bi H, Musiychuk K, de la Rosa P, Goldschmidt L, et al. Plant-expressed HA as a seasonal influenza vaccine candidate. Vaccine. 2008;26:2930–4. doi: 10.1016/j.vaccine.2008.03.045.
    1. Shoji Y, Farrance CE, Bi H, Shamloul M, Green B, Manceva S, et al. Immunogenicity of hemagglutinin from A/Bar-headed Goose/Qinghai/1A/05 and A/Anhui/1/05 strains of H5N1 influenza viruses produced in Nicotiana benthamiana plants. Vaccine. 2009;27:3467–70. doi: 10.1016/j.vaccine.2009.01.051.
    1. Shoji Y, Bi H, Musiychuk K, Rhee A, Horsey A, Roy G, et al. Plant-derived hemagglutinin protects ferrets against challenge infection with the A/Indonesia/05/05 strain of avian influenza. Vaccine. 2009;27:1087–92. doi: 10.1016/j.vaccine.2008.11.108.
    1. Mett V, Musiychuk K, Bi H, Farrance CE, Horsey A, Ugulava N, et al. A plant-produced influenza subunit vaccine protects ferrets against virus challenge. Influenza Other Respi Viruses. 2008;2:33–40. doi: 10.1111/j.1750-2659.2008.00037.x.
    1. Cox RJ, Brokstad KA, Zuckerman MA, Wood JM, Haaheim LR, Oxford JS. An early humoral immune response in peripheral blood following parenteral inactivated influenza vaccination. Vaccine. 1994;12:993–9. doi: 10.1016/0264-410X(94)90334-4.
    1. Cox RJ, Madhun AS, Hauge S, Sjursen H, Major D, Kuhne M, et al. A phase I clinical trial of a PER.C6 cell grown influenza H7 virus vaccine. Vaccine. 2009;27:1889–97. doi: 10.1016/j.vaccine.2009.01.116.
    1. Moris P, van der Most R, Leroux-Roels I, Clement F, Dramé M, Hanon E, et al. H5N1 influenza vaccine formulated with AS03 A induces strong cross-reactive and polyfunctional CD4 T-cell responses. J Clin Immunol. 2011;31:443–54. doi: 10.1007/s10875-010-9490-6.
    1. Roman F, Clément F, Dewé W, Walravens K, Maes C, Willekens J, et al. Effect on cellular and humoral immune responses of the AS03 adjuvant system in an A/H1N1/2009 influenza virus vaccine administered to adults during two randomized controlled trials. Clin Vaccine Immunol. 2011;18:835–43. doi: 10.1128/CVI.00480-10.
    1. Seder RA, Darrah PA, Roederer M. T-cell quality in memory and protection: implications for vaccine design. Nat Rev Immunol. 2008;8:247–58. doi: 10.1038/nri2274.
    1. Darrah PA, Patel DT, De Luca PM, Lindsay RWB, Davey DF, Flynn BJ, et al. Multifunctional TH1 cells define a correlate of vaccine-mediated protection against Leishmania major. Nat Med. 2007;13:843–50. doi: 10.1038/nm1592.
    1. Forbes EK, Sander C, Ronan EO, McShane H, Hill AVS, Beverley PCL, et al. Multifunctional, high-level cytokine-producing Th1 cells in the lung, but not spleen, correlate with protection against Mycobacterium tuberculosis aerosol challenge in mice. J Immunol. 2008;181:4955–64.
    1. Madhun AS, Haaheim LR, Nilsen MV, Cox RJ. Intramuscular Matrix-M-adjuvanted virosomal H5N1 vaccine induces high frequencies of multifunctional Th1 CD4+ cells and strong antibody responses in mice. Vaccine. 2009;27:7367–76. doi: 10.1016/j.vaccine.2009.09.044.
    1. Madhun AS, Haaheim LR, Nøstbakken JK, Ebensen T, Chichester J, Yusibov V, et al. Intranasal c-di-GMP-adjuvanted plant-derived H5 influenza vaccine induces multifunctional Th1 CD4+ cells and strong mucosal and systemic antibody responses in mice. Vaccine. 2011;29:4973–82. doi: 10.1016/j.vaccine.2011.04.094.
    1. Pedersen G, Halstensen A, Sjursen H, Naess A, Kristoffersen EK, Cox RJ. Pandemic influenza vaccination elicits influenza-specific CD4+ Th1-cell responses in hypogammaglobulinaemic patients: four case reports. Scand J Immunol. 2011;74:210–8. doi: 10.1111/j.1365-3083.2011.02561.x.
    1. D’Aoust M-A, Lavoie P-O, Couture MM-J, Trépanier S, Guay J-M, Dargis M, et al. Influenza virus-like particles produced by transient expression in Nicotiana benthamiana induce a protective immune response against a lethal viral challenge in mice. Plant Biotechnol J. 2008;6:930–40. doi: 10.1111/j.1467-7652.2008.00384.x.
    1. Landry N, Ward BJ, Trépanier S, Montomoli E, Dargis M, Lapini G, et al. Preclinical and clinical development of plant-made virus-like particle vaccine against avian H5N1 influenza. PLoS One. 2010;5:e15559. doi: 10.1371/journal.pone.0015559.
    1. Chu DW-S, Hwang S-J, Lim FS, Oh HML, Thongcharoen P, Yang P-C, et al. H5N1 Flu Study Group for Hong Kong, Singapore, Taiwan and Thailand Immunogenicity and tolerability of an AS03(A)-adjuvanted prepandemic influenza vaccine: a phase III study in a large population of Asian adults. Vaccine. 2009;27:7428–35. doi: 10.1016/j.vaccine.2009.07.102.
    1. Roman F, Vaman T, Gerlach B, Markendorf A, Gillard P, Devaster J-M. Immunogenicity and safety in adults of one dose of influenza A H1N1v 2009 vaccine formulated with and without AS03A-adjuvant: preliminary report of an observer-blind, randomised trial. Vaccine. 2010;28:1740–5. doi: 10.1016/j.vaccine.2009.12.014.
    1. Lund JM, Alexopoulou L, Sato A, Karow M, Adams NC, Gale NW, et al. Recognition of single-stranded RNA viruses by Toll-like receptor 7. Proc Natl Acad Sci U S A. 2004;101:5598–603. doi: 10.1073/pnas.0400937101.
    1. Diebold SS, Kaisho T, Hemmi H, Akira S, Reis e Sousa C. Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science. 2004;303:1529–31. doi: 10.1126/science.1093616.
    1. Dupuis M, Denis-Mize K, LaBarbara A, Peters W, Charo IF, McDonald DM, et al. Immunization with the adjuvant MF59 induces macrophage trafficking and apoptosis. Eur J Immunol. 2001;31:2910–8. doi: 10.1002/1521-4141(2001010)31:10<2910::AID-IMMU2910>;2-3.
    1. Seubert A, Monaci E, Pizza M, O’Hagan DT, Wack A. The adjuvants aluminum hydroxide and MF59 induce monocyte and granulocyte chemoattractants and enhance monocyte differentiation toward dendritic cells. J Immunol. 2008;180:5402–12.
    1. Mosca F, Tritto E, Muzzi A, Monaci E, Bagnoli F, Iavarone C, et al. Molecular and cellular signatures of human vaccine adjuvants. Proc Natl Acad Sci U S A. 2008;105:10501–6. doi: 10.1073/pnas.0804699105.
    1. Morel S, Didierlaurent A, Bourguignon P, Delhaye S, Baras B, Jacob V, et al. Adjuvant System AS03 containing α-tocopherol modulates innate immune response and leads to improved adaptive immunity. Vaccine. 2011;29:2461–73. doi: 10.1016/j.vaccine.2011.01.011.
    1. Galli G, Medini D, Borgogni E, Zedda L, Bardelli M, Malzone C, et al. Adjuvanted H5N1 vaccine induces early CD4+ T cell response that predicts long-term persistence of protective antibody levels. Proc Natl Acad Sci U S A. 2009;106:3877–82. doi: 10.1073/pnas.0813390106.
    1. Stephenson I, Bugarini R, Nicholson KG, Podda A, Wood JM, Zambon MC, et al. Cross-reactivity to highly pathogenic avian influenza H5N1 viruses after vaccination with nonadjuvanted and MF59-adjuvanted influenza A/Duck/Singapore/97 (H5N3) vaccine: a potential priming strategy. J Infect Dis. 2005;191:1210–5. doi: 10.1086/428948.
    1. Leroux-Roels I, Borkowski A, Vanwolleghem T, Dramé M, Clement F, Hons E, et al. Antigen sparing and cross-reactive immunity with an adjuvanted rH5N1 prototype pandemic influenza vaccine: a randomised controlled trial. Lancet. 2007;370:580–9. doi: 10.1016/S0140-6736(07)61297-5.
    1. Bernstein DI, Edwards KM, Dekker CL, Belshe R, Talbot HKB, Graham IL, et al. Effects of adjuvants on the safety and immunogenicity of an avian influenza H5N1 vaccine in adults. J Infect Dis. 2008;197:667–75. doi: 10.1086/527489.
    1. Levie K, Leroux-Roels I, Hoppenbrouwers K, Kervyn A-D, Vandermeulen C, Forgus S, et al. An adjuvanted, low-dose, pandemic influenza A (H5N1) vaccine candidate is safe, immunogenic, and induces cross-reactive immune responses in healthy adults. J Infect Dis. 2008;198:642–9. doi: 10.1086/590913.
    1. Cox RJ, Pedersen G, Madhun AS, Svindland S, Sævik M, Breakwell L, et al. Evaluation of a virosomal H5N1 vaccine formulated with Matrix M™ adjuvant in a phase I clinical trial. Vaccine. 2011;29:8049–59. doi: 10.1016/j.vaccine.2011.08.042.
    1. Karaolis DKR, Means TK, Yang D, Takahashi M, Yoshimura T, Muraille E, et al. Bacterial c-di-GMP is an immunostimulatory molecule. J Immunol. 2007;178:2171–81.
    1. Chen W, Kuolee R, Yan H. The potential of 3′,5′-cyclic diguanylic acid (c-di-GMP) as an effective vaccine adjuvant. Vaccine. 2010;28:3080–5. doi: 10.1016/j.vaccine.2010.02.081.
    1. Schild GC, Pereira MS, Chakraverty P. Single-radial-hemolysis: a new method for the assay of antibody to influenza haemagglutinin. Applications for diagnosis and seroepidemiologic surveillance of influenza. Bull World Health Organ. 1975;52:43–50.
    1. Brokstad KA, Cox RJ, Olofsson J, Jonsson R, Haaheim LR. Parenteral influenza vaccination induces a rapid systemic and local immune response. J Infect Dis. 1995;171:198–203. doi: 10.1093/infdis/171.1.198.
    1. Duvall MG, Precopio ML, Ambrozak DA, Jaye A, McMichael AJ, Whittle HC, et al. Polyfunctional T cell responses are a hallmark of HIV-2 infection. Eur J Immunol. 2008;38:350–63. doi: 10.1002/eji.200737768.

Source: PubMed

3
Prenumerera