First-in-Human Evaluation of the Safety and Immunogenicity of an Intranasally Administered Replication-Competent Sendai Virus-Vectored HIV Type 1 Gag Vaccine: Induction of Potent T-Cell or Antibody Responses in Prime-Boost Regimens

Julien Nyombayire, Omu Anzala, Brian Gazzard, Etienne Karita, Philip Bergin, Peter Hayes, Jakub Kopycinski, Gloria Omosa-Manyonyi, Akil Jackson, Jean Bizimana, Bashir Farah, Eddy Sayeed, Christopher L Parks, Makoto Inoue, Takashi Hironaka, Hiroto Hara, Tsugumine Shu, Tetsuro Matano, Len Dally, Burc Barin, Harriet Park, Jill Gilmour, Angela Lombardo, Jean-Louis Excler, Patricia Fast, Dagna S Laufer, Josephine H Cox, S001 Study Team, Rosine Ingabire, Gina Ouattara, Alan Steele, Anne Gumbe, Kundai Chinyenze, Sabrina Welsh, Carl Verlinde, Deborah King, Cynthia Bishop, Paramesh Chetty, Lorna Clark, Mumtaz Booley, Devika Zachariah, Kristen Syvertsen, Kamaal Anas, Marloes Naarding, Emmanuel Cormier, Jim Ackland, Mamoru Hasegawa, Julien Nyombayire, Omu Anzala, Brian Gazzard, Etienne Karita, Philip Bergin, Peter Hayes, Jakub Kopycinski, Gloria Omosa-Manyonyi, Akil Jackson, Jean Bizimana, Bashir Farah, Eddy Sayeed, Christopher L Parks, Makoto Inoue, Takashi Hironaka, Hiroto Hara, Tsugumine Shu, Tetsuro Matano, Len Dally, Burc Barin, Harriet Park, Jill Gilmour, Angela Lombardo, Jean-Louis Excler, Patricia Fast, Dagna S Laufer, Josephine H Cox, S001 Study Team, Rosine Ingabire, Gina Ouattara, Alan Steele, Anne Gumbe, Kundai Chinyenze, Sabrina Welsh, Carl Verlinde, Deborah King, Cynthia Bishop, Paramesh Chetty, Lorna Clark, Mumtaz Booley, Devika Zachariah, Kristen Syvertsen, Kamaal Anas, Marloes Naarding, Emmanuel Cormier, Jim Ackland, Mamoru Hasegawa

Abstract

Background: We report the first-in-human safety and immunogenicity assessment of a prototype intranasally administered, replication-competent Sendai virus (SeV)-vectored, human immunodeficiency virus type 1 (HIV-1) vaccine.

Methods: Sixty-five HIV-1-uninfected adults in Kenya, Rwanda, and the United Kingdom were assigned to receive 1 of 4 prime-boost regimens (administered at 0 and 4 months, respectively; ratio of vaccine to placebo recipients, 12:4): priming with a lower-dose SeV-Gag given intranasally, followed by boosting with an adenovirus 35-vectored vaccine encoding HIV-1 Gag, reverse transcriptase, integrase, and Nef (Ad35-GRIN) given intramuscularly (SLA); priming with a higher-dose SeV-Gag given intranasally, followed by boosting with Ad35-GRIN given intramuscularly (SHA); priming with Ad35-GRIN given intramuscularly, followed by boosting with a higher-dose SeV-Gag given intranasally (ASH); and priming and boosting with a higher-dose SeV-Gag given intranasally (SHSH).

Results: All vaccine regimens were well tolerated. Gag-specific IFN-γ enzyme-linked immunospot-determined response rates and geometric mean responses were higher (96% and 248 spot-forming units, respectively) in groups primed with SeV-Gag and boosted with Ad35-GRIN (SLA and SHA) than those after a single dose of Ad35-GRIN (56% and 54 spot-forming units, respectively) or SeV-Gag (55% and 59 spot-forming units, respectively); responses persisted for ≥8 months after completion of the prime-boost regimen. Functional CD8+ T-cell responses with greater breadth, magnitude, and frequency in a viral inhibition assay were also seen in the SLA and SHA groups after Ad35-GRIN boost, compared with those who received either vaccine alone. SeV-Gag did not boost T-cell counts in the ASH group. In contrast, the highest Gag-specific antibody titers were seen in the ASH group. Mucosal antibody responses were sporadic.

Conclusions: SeV-Gag primed functional, durable HIV-specific T-cell responses and boosted antibody responses. The prime-boost sequence appears to determine which arm of the immune response is stimulated.

Clinical trials registration: NCT01705990.

Keywords: HIV-1 vaccine; Sendai virus vector; adenovirus 35; immunogenicity; intranasal delivery; mucosal responses; prime-boost; replication competent.

© The Author 2016. Published by Oxford University Press for the Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
Administration of a Sendai virus (SeV)–vectored vaccine encoding human immunodeficiency virus 1 (HIV-1) Gag (SeV-Gag) primes Gag-specific T-cell responses detected by interferon γ enzyme-linked immunospot analysis. The y-axis shows spot-forming units (SFU)/106 peripheral blood mononuclear cells (PBMCs) on a log scale. All responses reflect subtraction of background spots. Black circles denote response below the cutoff, defined in the Materials and Methods, to the Gag peptide pool; red circles denote response above the cutoff to the Gag peptide pool. The overlaid box plots summarize the responses (ie, median value, the 1st and 3rd quartiles, and the 5th and 95th percentiles). Red bars represent median values. The placebo responses are combined for all groups. The prime-boost regimens as follows: priming with a lower-dose SeV-Gag given intranasally, followed by boosting with an adenovirus 35–vectored vaccine encoding HIV-1 Gag, reverse transcriptase, integrase, and Nef (Ad35-GRIN) given intramuscularly (SLA); priming with a higher-dose SeV-Gag given intranasally, followed by boosting with Ad35-GRIN given intramuscularly (SHA); priming with Ad35-GRIN given intramuscularly, followed by boosting with a higher-dose SeV-Gag given intranasally (ASH); and priming and boosting with a higher-dose SeV-Gag given intranasally (SHSH). Arrows show the timing of vaccinations in each group. In the x-axis, weeks represent the time after the most recent vaccination, to facilitate cross-regimen comparisons.
Figure 2.
Figure 2.
Characterization of human immunodeficiency virus (HIV-1) Gag–specific CD4+ and CD8+ T-cell responses. Gag-specific CD4+ and CD8+ T-cell responses as assessed by intracellular cytokine staining (ICS; A and B). Time points displayed are 2 weeks after the indicated vaccination. T-cell responses were evaluated by 7-color ICS to assess the expression of interleukin 2 (IL-2), interferon γ (IFN-γ), and tumor necrosis factor α (TNF-α) with peptides matched to the Gag peptide pool. The percentage of T cells expressing at least 1 cytokine (IL-2, IFN-γ, or TNF-α) is shown. Boxes represent interquartile ranges (IQRs), and whiskers extend to the 5th and 95th percentiles. Red bars represent median values. The placebo responses are combined for all groups. For the polyfunctional responses (bottom panels C and D), each dot represents a single volunteer. The prime-boost regimens as follows: priming with a lower-dose SeV-Gag given intranasally, followed by boosting with an adenovirus 35–vectored vaccine encoding HIV-1 Gag, reverse transcriptase, integrase, and Nef (Ad35-GRIN) given intramuscularly (SLA; red dots); priming with a higher-dose SeV-Gag given intranasally, followed by boosting with Ad35-GRIN given intramuscularly (SHA; green dots); priming with Ad35-GRIN given intramuscularly, followed by boosting with a higher-dose SeV-Gag given intranasally (ASH; orange dots); and priming and boosting with a higher-dose SeV-Gag given intranasally. Ad35-GRIN prime alone is represented by blue dots, and placebo is represented by purple dots.
Figure 3.
Figure 3.
Sendai virus (SeV)–vectored vaccine encoding human immunodeficiency virus 1 (HIV-1) Gag (SeV-Gag) enhances the breadth of inhibition of a panel of diverse HIV-1 isolates. The breadth of inhibition among 8 viruses was assessed at baseline (bl) and specified time points 2 weeks after the indicated vaccination (vac). Lines represent median values, whiskers represent the 1st and 3rd quartiles, and gray dots represent individual responses. The placebo (Pbo) responses are combined for all groups. The prime-boost regimens as follows: priming with a lower-dose SeV-Gag given intranasally, followed by boosting with an adenovirus 35–vectored vaccine encoding HIV-1 Gag, reverse transcriptase, integrase, and Nef (Ad35-GRIN) given intramuscularly (SLA); priming with a higher-dose SeV-Gag given intranasally, followed by boosting with Ad35-GRIN given intramuscularly (SHA); priming with Ad35-GRIN given intramuscularly, followed by boosting with a higher-dose SeV-Gag given intranasally (ASH).
Figure 4.
Figure 4.
Human immunodeficiency virus type 1 (HIV-1) Gag p24 immunoglobulin G (IgG) geometric mean titers (GMTs) after the following prime-boost combinations: priming with a lower-dose SeV-Gag given intranasally, followed by boosting with an adenovirus 35–vectored vaccine encoding HIV-1 Gag, reverse transcriptase, integrase, and Nef (Ad35-GRIN) given intramuscularly (SLA); priming with a higher-dose SeV-Gag given intranasally, followed by boosting with Ad35-GRIN given intramuscularly (SHA); priming with Ad35-GRIN given intramuscularly, followed by boosting with a higher-dose SeV-Gag given intranasally (ASH); and priming and boosting with a higher-dose SeV-Gag given intranasally (SHSH). Titers below the level of detection (ie, <100) were given a value of 50. Arrows indicate vaccination time points for each group. Placebo responses are combined for all groups. Abbreviation: CI, confidence interval.

References

    1. Fauci AS, Folkers GK, Marston HD. Ending the global HIV/AIDS pandemic: the critical role of an HIV vaccine. Clin Infect Dis 2014; 59(suppl 2):S80–4.
    1. Koff WC, Burton DR, Johnson PR et al. . Accelerating next-generation vaccine development for global disease prevention. Science 2013; 340:1232910.
    1. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S et al. . Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med 2009; 361:2209–20.
    1. Baden LR, Karita E, Mutua G et al. . Assessment of the safety and immunogenicity of 2 novel vaccine platforms for HIV-1 prevention: a randomized trial. Ann Intern Med 2016; 164:313–22.
    1. Goepfert PA, Elizaga ML, Seaton K et al. . Specificity and 6-month durability of immune responses induced by DNA and recombinant modified vaccinia Ankara vaccines expressing HIV-1 virus-like particles. J Infect Dis 2014; 210:99–110.
    1. Kalams SA, Parker SD, Elizaga M et al. . Safety and comparative immunogenicity of an HIV-1 DNA vaccine in combination with plasmid interleukin 12 and impact of intramuscular electroporation for delivery. J Infect Dis 2013; 208:818–29.
    1. Mpendo J, Mutua G, Nyombayire J et al. . A phase I double blind, placebo-controlled, randomized study of the safety and immunogenicity of electroporated HIV DNA with or without interleukin 12 in prime-boost combinations with an Ad35 HIV vaccine in healthy HIV-seronegative African adults. PLoS One 2015; 10:e0134287.
    1. Omosa-Manyonyi G, Mpendo J, Ruzagira E et al. . A phase I double blind, placebo-controlled, randomized study of the safety and immunogenicity of an adjuvanted HIV-1 Gag-Pol-Nef fusion protein and adenovirus 35 Gag-RT-Int-Nef vaccine in healthy HIV-uninfected African adults. PLoS One 2015; 10:e0125954.
    1. Belshe RB, Edwards KM, Vesikari T et al. . Live attenuated versus inactivated influenza vaccine in infants and young children. N Engl J Med 2007; 356:685–96.
    1. Englund JA, Karron RA, Cunningham CK et al. . Safety and infectivity of two doses of live-attenuated recombinant cold-passaged human parainfluenza type 3 virus vaccine rHPIV3cp45 in HPIV3-seronegative young children. Vaccine 2013; 31:5706–12.
    1. Malkin E, Yogev R, Abughali N et al. . Safety and immunogenicity of a live attenuated RSV vaccine in healthy RSV-seronegative children 5 to 24 months of age. PLoS One 2013; 8:e77104.
    1. Yuki Y, Kiyono H. Mucosal vaccines: novel advances in technology and delivery. Expert Rev Vaccines 2009; 8:1083–97.
    1. Wright PF, Mestecky J, McElrath MJ et al. . Comparison of systemic and mucosal delivery of 2 canarypox virus vaccines expressing either HIV-1 genes or the gene for rabies virus G protein. J Infect Dis 2004; 189:1221–31.
    1. Russell CJ, Hurwitz JL. Sendai virus as a backbone for vaccines against RSV and other human paramyxoviruses. Expert Rev Vaccines 2016; 15:189–200.
    1. Seki S, Matano T. Development of a Sendai virus vector-based AIDS vaccine inducing T cell responses. Expert Rev Vaccines 2016; 15:119–27.
    1. Skiadopoulos MH, Surman SR, Riggs JM et al. . Sendai virus, a murine parainfluenza virus type 1, replicates to a level similar to human PIV1 in the upper and lower respiratory tract of African green monkeys and chimpanzees. Virology 2002; 297:153–60.
    1. Yonemitsu Y, Kitson C, Ferrari S et al. . Efficient gene transfer to airway epithelium using recombinant Sendai virus. Nat Biotechnol 2000; 18:970–3.
    1. Gorman WL, Gill DS, Scroggs RA, Portner A. The hemagglutinin-neuraminidase glycoproteins of human parainfluenza virus type 1 and Sendai virus have high structure-function similarity with limited antigenic cross-reactivity. Virology 1990; 175:211–21.
    1. Lyn D, Gill DS, Scroggs RA, Portner A. The nucleoproteins of human parainfluenza virus type 1 and Sendai virus share amino acid sequences and antigenic and structural determinants. J Gen Virol 1991; 72(Pt 4):983–7.
    1. Power UF, Ryan KW, Portner A. Sequence characterization and expression of the matrix protein gene of human parainfluenza virus type 1. Virology 1992; 191:947–52.
    1. Takimoto T, Bousse T, Portner A. Molecular cloning and expression of human parainfluenza virus type 1 L gene. Virus Res 2000; 70:45–53.
    1. Adderson E, Branum K, Sealy RE et al. . Safety and immunogenicity of an intranasal Sendai virus-based human parainfluenza virus type 1 vaccine in 3- to 6-year-old children. Clin Vaccine Immunol 2015; 22:298–303.
    1. Slobod KS, Shenep JL, Lujan-Zilbermann J et al. . Safety and immunogenicity of intranasal murine parainfluenza virus type 1 (Sendai virus) in healthy human adults. Vaccine 2004; 22:3182–6.
    1. Hara H, Hara H, Hironaka T et al. . Prevalence of specific neutralizing antibodies against Sendai virus in populations from different geographic areas: implications for AIDS vaccine development using Sendai virus vectors. Hum Vaccin 2011; 7:639–45.
    1. Demberg T, Robert-Guroff M. Mucosal immunity and protection against HIV/SIV infection: strategies and challenges for vaccine design. Int Rev Immunol 2009; 28:20–48.
    1. Poles J, Alvarez Y, Hioe CE. Induction of intestinal immunity by mucosal vaccines as a means of controlling HIV infection. AIDS Res Hum Retroviruses 2014; 30:1027–40.
    1. Kano M, Matano T, Nakamura H et al. . Elicitation of protective immunity against simian immunodeficiency virus infection by a recombinant Sendai virus expressing the Gag protein. AIDS 2000; 14:1281–2.
    1. Matano T, Kano M, Nakamura H, Takeda A, Nagai Y. Rapid appearance of secondary immune responses and protection from acute CD4 depletion after a highly pathogenic immunodeficiency virus challenge in macaques vaccinated with a DNA prime/Sendai virus vector boost regimen. J Virol 2001; 75:11891–6.
    1. Matano T, Kobayashi M, Igarashi H et al. . Cytotoxic T lymphocyte-based control of simian immunodeficiency virus replication in a preclinical AIDS vaccine trial. J Exp Med 2004; 199:1709–18.
    1. Moriya C, Horiba S, Kurihara K et al. . Intranasal Sendai viral vector vaccination is more immunogenic than intramuscular under pre-existing anti-vector antibodies. Vaccine 2011; 29:8557–63.
    1. Keefer MC, Gilmour J, Hayes P et al. . A phase I double blind, placebo-controlled, randomized study of a multigenic HIV-1 adenovirus subtype 35 vector vaccine in healthy uninfected adults. PLoS One 2012; 7:e41936.
    1. Stiles T, Grant V, Mawbey T. Good clinical laboratory practice (GCLP). A quality system for laboratories that undertake the analysis of samples from clinical trials. Ipswich, UK: BARQA, 2010.
    1. Kato A, Sakai Y, Shioda T, Kondo T, Nakanishi M, Nagai Y. Initiation of Sendai virus multiplication from transfected cDNA or RNA with negative or positive sense. Genes Cells 1996; 1:569–79.
    1. Hasan MK, Kato A, Shioda T, Sakai Y, Yu D, Nagai Y. Creation of an infectious recombinant Sendai virus expressing the firefly luciferase gene from the 3′ proximal first locus. J Gen Virol 1997; 78(Pt 11):2813–20.
    1. Mutua G, Farah B, Langat R, et al; Hiv-Core 004 Study Group T. Broad HIV-1 inhibition in vitro by vaccine-elicited CD8(+) T cells in African adults. Mol Ther Methods Clin Dev 2016; 3:16061.
    1. Bergin PJ, Langat R, Omosa-Manyonyi G et al. . Assessment of anti-HIV-1 antibodies in oral and nasal compartments of volunteers from 3 different populations. J Acquir Immune Defic Syndr 2016; 73:130–7.
    1. Omosa-Manyonyi G, Park H, Mutua G et al. . Acceptability and feasibility of repeated mucosal specimen collection in clinical trial participants in Kenya. PLoS One 2014; 9:e110228.
    1. Kaltsidis H, Cheeseman H, Kopycinski J et al. . Measuring human T cell responses in blood and gut samples using qualified methods suitable for evaluation of HIV vaccine candidates in clinical trials. J Immunol Methods 2011; 370:43–54.
    1. Shacklett BL, Yang O, Hausner MA et al. . Optimization of methods to assess human mucosal T-cell responses to HIV infection. J Immunol Methods 2003; 279:17–31.
    1. Spentzou A, Bergin P, Gill D et al. . Viral inhibition assay: a CD8 T cell neutralization assay for use in clinical trials of HIV-1 vaccine candidates. J Infect Dis 2010; 201:720–9.
    1. Hayes PJ, Cox JH, Coleman AR et al. . Adenovirus-based HIV-1 vaccine candidates tested in efficacy trials elicit CD8+ T cells with limited breadth of HIV-1 inhibition. AIDS 2016; 30:1703–12.
    1. Croyle MA, Patel A, Tran KN et al. . Nasal delivery of an adenovirus-based vaccine bypasses pre-existing immunity to the vaccine carrier and improves the immune response in mice. PLoS One 2008; 3:e3548.
    1. De Rosa SC, Thomas EP, Bui J et al. . HIV-DNA priming alters T cell responses to HIV-adenovirus vaccine even when responses to DNA are undetectable. J Immunol 2011; 187:3391–401.
    1. Lhomme E, Richert L, Moodie Z et al. . Early CD4+ T cell responses are associated with subsequent CD8+ T cell responses to an rAd5-based prophylactic prime-boost HIV vaccine strategy. PLoS One 2016; 11:e0152952.
    1. Churchyard GJ, Morgan C, Adams E et al. . A phase IIA randomized clinical trial of a multiclade HIV-1 DNA prime followed by a multiclade rAd5 HIV-1 vaccine boost in healthy adults (HVTN204). PLoS One 2011; 6:e21225.
    1. Fuchs JD, Bart PA, Frahm N et al. . Safety and immunogenicity of a recombinant adenovirus serotype 35-vectored HIV-1 vaccine in adenovirus serotype 5 seronegative and seropositive individuals. J AIDS Clin Res 2015; 6:461.
    1. Graham BS, Koup RA, Roederer M et al. . Phase 1 safety and immunogenicity evaluation of a multiclade HIV-1 DNA candidate vaccine. J Infect Dis 2006; 194:1650–60.
    1. Jaoko W, Karita E, Kayitenkore K et al. . Safety and immunogenicity study of multiclade HIV-1 adenoviral vector vaccine alone or as boost following a multiclade HIV-1 DNA vaccine in Africa. PLoS One 2010; 5:e12873.
    1. Koup RA, Roederer M, Lamoreaux L et al. . Priming immunization with DNA augments immunogenicity of recombinant adenoviral vectors for both HIV-1 specific antibody and T-cell responses. PLoS One 2010; 5:e9015.
    1. Fuchs JD, Frank I, Elizaga ML et al. . First-in-human evaluation of the safety and immunogenicity of a recombinant vesicular stomatitis virus human immunodeficiency virus-1 gag vaccine (HVTN 090). Open Forum Infect Dis 2015; 2:ofv082.
    1. Kalams SA, Parker S, Jin X et al. . Safety and immunogenicity of an HIV-1 gag DNA vaccine with or without IL-12 and/or IL-15 plasmid cytokine adjuvant in healthy, HIV-1 uninfected adults. PLoS One 2012; 7:e29231.

Source: PubMed

3
구독하다