Human adenovirus 5-vectored Plasmodium falciparum NMRC-M3V-Ad-PfCA vaccine encoding CSP and AMA1 is safe, well-tolerated and immunogenic but does not protect against controlled human malaria infection

Cindy Tamminga, Martha Sedegah, Santina Maiolatesi, Charlotte Fedders, Sharina Reyes, Anatalio Reyes, Carlos Vasquez, Yolanda Alcorta, Ilin Chuang, Michele Spring, Michael Kavanaugh, Harini Ganeshan, Jun Huang, Maria Belmonte, Esteban Abot, Arnel Belmonte, Joglenna Banania, Fouzia Farooq, Jittawadee Murphy, Jack Komisar, Nancy O Richie, Jason Bennett, Keith Limbach, Noelle B Patterson, Joseph T Bruder, Meng Shi, Edward Miller, Sheetij Dutta, Carter Diggs, Lorraine A Soisson, Michael R Hollingdale, Judith E Epstein, Thomas L Richie, Cindy Tamminga, Martha Sedegah, Santina Maiolatesi, Charlotte Fedders, Sharina Reyes, Anatalio Reyes, Carlos Vasquez, Yolanda Alcorta, Ilin Chuang, Michele Spring, Michael Kavanaugh, Harini Ganeshan, Jun Huang, Maria Belmonte, Esteban Abot, Arnel Belmonte, Joglenna Banania, Fouzia Farooq, Jittawadee Murphy, Jack Komisar, Nancy O Richie, Jason Bennett, Keith Limbach, Noelle B Patterson, Joseph T Bruder, Meng Shi, Edward Miller, Sheetij Dutta, Carter Diggs, Lorraine A Soisson, Michael R Hollingdale, Judith E Epstein, Thomas L Richie

Abstract

Background: In a prior study, a DNA prime / adenovirus boost vaccine (DNA/Ad) expressing P. falciparum circumsporozoite protein (CSP) and apical membrane antigen-1 (AMA1) (NMRC-M3V-D/Ad-PfCA Vaccine) induced 27% protection against controlled human malaria infection (CHMI). To investigate the contribution of DNA priming, we tested the efficacy of adenovirus vaccine alone (NMRC-M3V-Ad-PfCA ) in a Phase 1 clinical trial.

Methodology/principal findings: The regimen was a single intramuscular injection with two non-replicating human serotype 5 adenovectors encoding CSP and AMA1, respectively. One x 10 (10) particle units of each construct were combined prior to administration. The regimen was safe and well-tolerated. Four weeks later, 18 study subjects received P. falciparum CHMI administered by mosquito bite. None were fully protected although one showed delayed onset of parasitemia. Antibody responses were low, with geometric mean CSP ELISA titer of 381 (range<50-1626) and AMA1 ELISA of 4.95 µg/mL (range 0.2-38). Summed ex vivo IFN-γ ELISpot responses to overlapping peptides were robust, with geometric mean spot forming cells/million peripheral blood mononuclear cells [sfc/m] for CSP of 273 (range 38-2550) and for AMA1 of 1303 (range 435-4594). CD4+ and CD8+ T cell IFN-γ responses to CSP were positive by flow cytometry in 25% and 56% of the research subjects, respectively, and to AMA1 in 94% and 100%, respectively.

Significance: In contrast to DNA/Ad, Ad alone did not protect against CHMI despite inducing broad, cell-mediated immunity, indicating that DNA priming is required for protection by the adenovirus-vectored vaccine. ClinicalTrials.gov Identifier: NCT00392015.

Trial registration: ClinicalTrials.gov NCT00392015 NCT00870987.

Keywords: T cells; adenovirus; antibody; efficacy; malaria; vaccine.

Figures

https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3906401/bin/hvi-9-2165-g1.jpg
Figure 1. Schematic of Adenovirus CSP and AMA1 vaccines. Each panel presents the native protein (top of each panel) and the protein expressed by the Ad construct (bottom of each panel) for the CSP (A) and AMA1 (B) vaccine antigens. N = N-terminus; C = carboxy terminus; TM = transmembrane domain. Identical colors indicate identical sequences.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3906401/bin/hvi-9-2165-g2.jpg
Figure 2. Trial design. Subjects were immunized week 0 and challenged week 4. Samples for measuring cell-mediated immunity (ELISpot assay and flow cytometry) and antibody levels (ELISA and IFA) were collected at six time points (black arrows): Pre (pre-immunization), Post-Ad (*22–23 d after immunization), Post-Ch+4 (four weeks after challenge), Post-Ch+12 (12 weeks after challenge), Post-Ch+20 (20 weeks after challenge), and Post-Ch+48 (48 weeks after challenge).
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3906401/bin/hvi-9-2165-g3.jpg
Figure 3. Flow diagram of immunized and control volunteers. Thirty-seven volunteers met all eligibility criteria of whom 11 withdrew consent, and 26 volunteers were allocated to the immunization group (n = 20) and infectivity controls (n = 6) respectively. Two immunized volunteers were not challenged due to family reasons and poor compliance, respectively. Approximately two years after challenge, an infectivity control died of causes unrelated to the vaccine.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3906401/bin/hvi-9-2165-g4.jpg
Figure 4. Development of parasitemia in the immunized and infectivity volunteers. Parasitemia-free survival curves (Kaplan-Meier) for immunized volunteers and infectivity controls based on microscopic examination of peripheral blood smears.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3906401/bin/hvi-9-2165-g5.jpg
Figure 5. Antibody responses by ELISA to CSP and AMA1. Group geomean CSP and AMA1 ELISA activities for the 18 recipients were significantly higher than baseline (*) CSP: Post-Ad (CSP p = < 0.0001), Post-Ch+4 CSP (p = < 0.0001), Post-Ch+12 (CSP p = < 0.0001) and Post-Ch+20 (CSP p = 0.0003); AMA1: Post-Ad (p = < 0.0001), Post-Ch+4 CSP (p = < 0.0001), Post-Ch+12 (p = < 0.0001) and Post-Ch+20 (p = < 0.0001). For explanation of box plots (including outliers) see statistics section at the end of Methods below.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3906401/bin/hvi-9-2165-g6.jpg
Figure 6. Antibody responses by IFA to P. falciparum sporozoites and asexual blood stages. Group geomean IFA activities were significantly higher than baseline (*): Sporozoites: Post-Ad (p = 0. < 0001), Post-Ch+4 (p = 0. < 0001), Post-Ch+12 (p = 0.0015), Post-Ch+20 (p = 0.0002); red blood stages: Post-Ad (p = < 0.0001), Post-Ch+4 (p = < 0.0001), and Post-Ch+12 (p = < 0.0001) and Post-Ch+20 (p = < 0.0001) (mixed linear model). For explanation of box plots (including outliers) see statistics section at the end of Methods below.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3906401/bin/hvi-9-2165-g7.jpg
Figure 7. Ex vivo T cell IFN-γ activities by ELISpot Assay for CSP and AMA1. Group geomean IFA activities were significantly higher than baseline (*): CSP: Post-Ad (p = < 0.0001), Post-Ch+4 (p = < 0.0001), Post-Ch+12 (p = 0.0083), Post-Ch+20 (p = 0.0009) Post Ch+48 (p = < 0.0001; AMA1 Post-Ad (p = < 0.0001), Post-Ch+4 (p = < 0.0001), Post-Ch+12 (p = 0.0056), Post-Ch+20 (p = 0.0019), Post Ch+48 (p = < 0.0013). For explanation of box plots (including outliers) see statistics section at the end of Methods below.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3906401/bin/hvi-9-2165-g8.jpg
Figure 8. CD4+ and CD8+ T cell IFN-γ activities by flow cytometry for CSP and AMA1. The box plots represent IFN-γ-producing CD4+ or CD8+ T cell frequencies as percentage of gated CD4+ or CD8+ T cells, measured by flow cytometry assays after stimulation with a single CSP or AMA1 megapool containing all individual peptide pools for each antigen. Arrow denotes v156 who had very high CD8+ T cell activity to AMA1 (2.08%) at Post-Ad. IFN-γ-producing CD4+ T cell activities for CSP were only significantly higher than baseline (*) at Post-Ad (p = 0.0017); CD4+ T cell activities for AMA1 were significantly higher than baseline (*) Post-Ad (p = 0.0002), Post-Ch+4 (p = 0.0017), Post-Ch+20 (p = 0.0054) and Post-Ch+48 (p = 0.0035). IFN-γ-producing CD8+ T cell activities for CSP were only marginally higher than baseline (*) at Post Ad (p = 0.07); CD8+ T cell activities for AMA1 were significantly higher than baseline (*) at Post-Ad (p = 0.0021) and Post-Ch+4 (p = 0.017). For explanation of box plots (including outliers) see statistics section at the end of Methods below.

References

    1. Overstreet MG, Cockburn IA, Chen YC, Zavala F. Protective CD8 T cells against Plasmodium liver stages: immunobiology of an ‘unnatural’ immune response. Immunol Rev. 2008;225:272–83. doi: 10.1111/j.1600-065X.2008.00671.x.
    1. Butler NS, Schmidt NW, Vaughan AM, Aly AS, Kappe SH, Harty JT. Superior antimalarial immunity after vaccination with late liver stage-arresting genetically attenuated parasites. Cell Host Microbe. 2011;9:451–62. doi: 10.1016/j.chom.2011.05.008.
    1. Moorthy VS, Ballou WR. Immunological mechanisms underlying protection mediated by RTS,S: a review of the available data. Malar J. 2009;8:312. doi: 10.1186/1475-2875-8-312.
    1. Halstead SB, Thomas SJ. New Japanese encephalitis vaccines: alternatives to production in mouse brain. Expert Rev Vaccines. 2011;10:355–64. doi: 10.1586/erv.11.7.
    1. Sedegah M, Tamminga C, McGrath S, House B, Ganeshan H, Lejano J, et al. Adenovirus 5-vectored P. falciparum vaccine expressing CSP and AMA1. Part A: safety and immunogenicity in seronegative adults. PLoS One. 2011;6:e24586. doi: 10.1371/journal.pone.0024586.
    1. Sheehy SH, Duncan CJ, Elias SC, Choudhary P, Biswas S, Halstead FD, Collins KA, Edwards NJ, Douglas AD, Anagnostou NA, et al. ChAd63-MVA-vectored blood-stage malaria vaccines targeting MSP1 and AMA1: assessment of efficacy against mosquito bite challenge in humans. Mol Ther. 2012;20:2355–68. doi: 10.1038/mt.2012.223.
    1. Webster DP, Dunachie S, Vuola JM, Berthoud T, Keating S, Laidlaw SM, et al. Enhanced T cell-mediated protection against malaria in human challenges by using the recombinant poxviruses FP9 and modified vaccinia virus Ankara. Proc Natl Acad Sci U S A. 2005;102:4836–41. doi: 10.1073/pnas.0406381102.
    1. Wang R, Epstein J, Baraceros FM, Gorak EJ, Charoenvit Y, Carucci DJ, et al. Induction of CD4(+) T cell-dependent CD8(+) type 1 responses in humans by a malaria DNA vaccine. Proc Natl Acad Sci U S A. 2001;98:10817–22. doi: 10.1073/pnas.181123498.
    1. Wang R, Doolan DL, Charoenvit Y, Hedstrom RC, Gardner MJ, Hobart P, et al. Simultaneous induction of multiple antigen-specific cytotoxic T lymphocytes in nonhuman primates by immunization with a mixture of four Plasmodium falciparum DNA plasmids. Infect Immun. 1998;66:4193–202.
    1. Reyes-Sandoval A, Berthoud T, Alder N, Siani L, Gilbert SC, Nicosia A, et al. Prime-boost immunization with adenoviral and modified vaccinia virus Ankara vectors enhances the durability and polyfunctionality of protective malaria CD8+ T-cell responses. Infect Immun. 2010;78:145–53. doi: 10.1128/IAI.00740-09.
    1. Jiang G, Shi M, Conteh S, Richie N, Banania G, Geneshan H, et al. Sterile protection against Plasmodium knowlesi in rhesus monkeys from a malaria vaccine: comparison of heterologous prime boost strategies. PLoS One. 2009;4:e6559. doi: 10.1371/journal.pone.0006559.
    1. Sedegah M, Hedstrom R, Hobart P, Hoffman SL. Protection against malaria by immunization with plasmid DNA encoding circumsporozoite protein. Proc Natl Acad Sci U S A. 1994;91:9866–70. doi: 10.1073/pnas.91.21.9866.
    1. Le TP, Coonan KM, Hedstrom RC, Charoenvit Y, Sedegah M, Epstein JE, et al. Safety, tolerability and humoral immune responses after intramuscular administration of a malaria DNA vaccine to healthy adult volunteers. Vaccine. 2000;18:1893–901. doi: 10.1016/S0264-410X(99)00407-7.
    1. Epstein JE, Gorak EJ, Charoenvit Y, Wang R, Freydberg N, Osinowo O, et al. Safety, tolerability, and lack of antibody responses after administration of a PfCSP DNA malaria vaccine via needle or needle-free jet injection, and comparison of intramuscular and combination intramuscular/intradermal routes. Hum Gene Ther. 2002;13:1551–60. doi: 10.1089/10430340260201644.
    1. Richie TL, Charoenvit Y, Wang R, Epstein JE, Hedstrom RC, Kumar S, et al. Clinical trial in healthy malaria-naïve adults to evaluate the safety, tolerability, immunogenicity and efficacy of MuStDO5, a five-gene, sporozoite/hepatic stage Plasmodium falciparum DNA vaccine combined with escalating dose human GM-CSF DNA. Hum Vaccin Immunother. 2012;8:1564–84. doi: 10.4161/hv.22129.
    1. Chuang I, Sedegah M, Cicatelli S, Spring M, Polhemus M, Tamminga C, et al. DNA prime/adenovirus boost malaria vaccine encoding P. falciparum CSP and AMA1 induces sterile protection associated with cell-mediated immunity. PLoS ONE. 2013; 8:e55571
    1. Tamminga C, Sedegah M, Regis D, Chuang I, Epstein JE, Spring M, et al. Adenovirus-5-vectored P. falciparum vaccine expressing CSP and AMA1. Part B: safety, immunogenicity and protective efficacy of the CSP component. PLoS One. 2011;6:e25868. doi: 10.1371/journal.pone.0025868.
    1. Sprangers MC, Lakhai W, Koudstaal W, Verhoeven M, Koel BF, Vogels R, et al. Quantifying adenovirus-neutralizing antibodies by luciferase transgene detection: addressing preexisting immunity to vaccine and gene therapy vectors. J Clin Microbiol. 2003;41:5046–52. doi: 10.1128/JCM.41.11.5046-5052.2003.
    1. Chulay JD, Schneider I, Cosgriff TM, Hoffman SL, Ballou WR, Quakyi IA, et al. Malaria transmitted to humans by mosquitoes infected from cultured Plasmodium falciparum. Am J Trop Med Hyg. 1986;35:66–8.
    1. Sheehy SH, Duncan CJ, Elias SC, Biswas S, Collins KA, O’Hara GA, et al. Phase Ia clinical evaluation of the safety and immunogenicity of the Plasmodium falciparum blood-stage antigen AMA1 in ChAd63 and MVA vaccine vectors. PLoS One. 2012;7:e31208. doi: 10.1371/journal.pone.0031208.
    1. Good MF, Quakyi IA, Saul A, Berzofsky JA, Carter R, Miller LH. Human T clones reactive to the sexual stages of Plasmodium falciparum malaria. High frequency of gamete-reactive T cells in peripheral blood from nonexposed donors. J Immunol. 1987;138:306–11.
    1. Fell AH, Silins SL, Baumgarth N, Good MF. Plasmodium falciparum-specific T cell clones from non-exposed and exposed donors are highly diverse in TCR beta chain V segment usage. Int Immunol. 1996;8:1877–87. doi: 10.1093/intimm/8.12.1877.
    1. Holst PJ, Ørskov C, Thomsen AR, Christensen JP. Quality of the transgene-specific CD8+ T cell response induced by adenoviral vector immunization is critically influenced by virus dose and route of vaccination. J Immunol. 2010;184:4431–9. doi: 10.4049/jimmunol.0900537.
    1. De Rosa SC, Thomas EP, Bui J, Huang Y, deCamp A, Morgan C, et al. National Institute of Allergy and Infectious Diseases HIV Vaccine Trials Network HIV-DNA priming alters T cell responses to HIV-adenovirus vaccine even when responses to DNA are undetectable. J Immunol. 2011;187:3391–401. doi: 10.4049/jimmunol.1101421.
    1. Park JY, Jin DH, Lee CM, Jang MJ, Lee SY, Shin HS, et al. CD4+ TH1 cells generated by Ii-PADRE DNA at prime phase are important to induce effectors and memory CD8+ T cells. J Immunother. 2010;33:510–22. doi: 10.1097/CJI.0b013e3181d75cef.
    1. Reyes-Sandoval A, Wyllie DH, Bauza K, Milicic A, Forbes EK, Rollier CS, et al. CD8+ T effector memory cells protect against liver-stage malaria. J Immunol. 2011;187:1347–57. doi: 10.4049/jimmunol.1100302.
    1. Jobe O, Donofrio G, Sun G, Liepinsh D, Schwenk R, Krzych U. Immunization with radiation-attenuated Plasmodium berghei sporozoites induces liver cCD8alpha+DC that activate CD8+T cells against liver-stage malaria. PLoS One. 2009;4:e5075. doi: 10.1371/journal.pone.0005075.
    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. Gaziano TA, Young CR, Fitzmaurice G, Atwood S, Gaziano JM. Laboratory-based versus non-laboratory-based method for assessment of cardiovascular disease risk: the NHANES I Follow-up Study cohort. Lancet. 2008;371:923–31. doi: 10.1016/S0140-6736(08)60418-3.
    1. Harro CD, Robertson MN, Lally MA, O’Neill LD, Edupuganti S, Goepfert PA, et al. Merck V520-007/012 Study Teams Safety and immunogenicity of adenovirus-vectored near-consensus HIV type 1 clade B gag vaccines in healthy adults. AIDS Res Hum Retroviruses. 2009;25:103–14. doi: 10.1089/aid.2008.0212.
    1. Stoute JA, Slaoui M, Heppner DG, Momin P, Kester KE, Desmons P, et al. A preliminary evaluation of a recombinant circumsporozoite protein vaccine against Plasmodium falciparum malaria. RTS,S Malaria Vaccine Evaluation Group. N Engl J Med. 1997;336:86–91. doi: 10.1056/NEJM199701093360202.
    1. McGill R. Variations of Box Plots. Am Stat. 1978;32:12–6.

Source: PubMed

3
Tilaa