Adenovirus 5-vectored P. falciparum vaccine expressing CSP and AMA1. Part A: safety and immunogenicity in seronegative adults

Martha Sedegah, Cindy Tamminga, Shannon McGrath, Brent House, Harini Ganeshan, Jennylynn Lejano, Esteban Abot, Glenna J Banania, Renato Sayo, Fouzia Farooq, Maria Belmonte, Nalini Manohar, Nancy O Richie, Chloe Wood, Carole A Long, David Regis, Francis T Williams, Meng Shi, Ilin Chuang, Michele Spring, Judith E Epstein, Jose Mendoza-Silveiras, Keith Limbach, Noelle B Patterson, Joseph T Bruder, Denise L Doolan, C Richter King, Lorraine Soisson, Carter Diggs, Daniel Carucci, Sheetij Dutta, Michael R Hollingdale, Christian F Ockenhouse, Thomas L Richie, Martha Sedegah, Cindy Tamminga, Shannon McGrath, Brent House, Harini Ganeshan, Jennylynn Lejano, Esteban Abot, Glenna J Banania, Renato Sayo, Fouzia Farooq, Maria Belmonte, Nalini Manohar, Nancy O Richie, Chloe Wood, Carole A Long, David Regis, Francis T Williams, Meng Shi, Ilin Chuang, Michele Spring, Judith E Epstein, Jose Mendoza-Silveiras, Keith Limbach, Noelle B Patterson, Joseph T Bruder, Denise L Doolan, C Richter King, Lorraine Soisson, Carter Diggs, Daniel Carucci, Sheetij Dutta, Michael R Hollingdale, Christian F Ockenhouse, Thomas L Richie

Abstract

Background: Models of immunity to malaria indicate the importance of CD8+ T cell responses for targeting intrahepatic stages and antibodies for targeting sporozoite and blood stages. We designed a multistage adenovirus 5 (Ad5)-vectored Plasmodium falciparum malaria vaccine, aiming to induce both types of responses in humans, that was tested for safety and immunogenicity in a Phase 1 dose escalation trial in Ad5-seronegative volunteers.

Methodology/principal findings: The NMRC-M3V-Ad-PfCA vaccine combines two adenovectors encoding circumsporozoite protein (CSP) and apical membrane antigen-1 (AMA1). Group 1 (n = 6) healthy volunteers received one intramuscular injection of 2×10∧10 particle units (1×10∧10 each construct) and Group 2 (n = 6) a five-fold higher dose. Transient, mild to moderate adverse events were more pronounced with the higher dose. ELISpot responses to CSP and AMA1 peaked at 1 month, were higher in the low dose (geomean CSP = 422, AMA1 = 862 spot forming cells/million) than in the high dose (CSP = 154, p = 0.049, AMA1 = 423, p = 0.045) group and were still positive at 12 months in a number of volunteers. ELISpot depletion assays identified dependence on CD4+ or on both CD4+ and CD8+ T cells, with few responses dependent only on CD8+ T cells. Intracellular cytokine staining detected stronger CD8+ than CD4+ T cell IFN-γ responses (CSP p = 0.0001, AMA1 p = 0.003), but similar frequencies of multifunctional CD4+ and CD8+ T cells secreting two or more of IFN-γ, TNF-α or IL-2. Median fluorescence intensities were 7-10 fold higher in triple than single secreting cells. Antibody responses were low but trended higher in the high dose group and did not inhibit growth of cultured P. falciparum blood stage parasites.

Significance: As found in other trials, adenovectored vaccines appeared safe and well-tolerated at doses up to 1×10∧11 particle units. This is the first demonstration in humans of a malaria vaccine eliciting strong CD8+ T cell IFN-γ responses.

Trial registration: ClinicalTrials.govNCT00392015.

Conflict of interest statement

Competing Interests: The authors have read the journal's policy and have the following conflicts: SM NOR CW worked for Clinical Research Management; JTB CRK worked for GenVec; DLD held a Pfizer Australia Senior Research Fellowship; JTB CRK DLD DC KL are inventors listed on U.S. Patent No., U.S. Patent No. 2009-0148477 A1, and international patent application PCT/US06/33982, titled “Adenoviral Vector-based Malaria Vaccines”; JTB CRK TLR KL DLD. are inventors listed on U.S. Patent Application 12/522,335, and international patent application PCT/US08/50565 titled “Adenoviral Vector-based Malaria Vaccines”. These do not alter the authors' adherence to all PLoS ONE policies on sharing data and materials.

Figures

Figure 1. Schematic of Ad5-PfCSP and Ad5-PfAMA1…
Figure 1. Schematic of Ad5-PfCSP and Ad5-PfAMA1 vectors.
The parent adenovector was a serotype 5 adenovirus carrying deletions in E1, E4 and part of the E3 region with a transcriptionally inert spacer inserted into the site of the E4 deletion (TIS1), the resulting replication defective vector called GV11D. Codon-optimized CSP or AMA1 genes were inserted into the E1 region under the control of a cytomegalovirus promoter (hCMV IE). SV40 pA = simian virus 40 polyadenylation sequence.
Figure 2. Flow diagram of volunteers in…
Figure 2. Flow diagram of volunteers in Groups 1 and 2.
The first six volunteers were allocated to Group 1 and the subsequent six volunteers to Group 2. *Reasons for exclusion: lost to follow up (2), moved out of area (1), deployed (1), job commitments (1).
Figure 3. Neutrophil kinetics in Groups 1…
Figure 3. Neutrophil kinetics in Groups 1 and 2 after immunization.
A fall in neutrophil count was observed in most volunteers at 2–3 days post-immunization with return to normal or near normal levels by day 7 post-immunization.
Figure 4. ELISpot activity of serial bleeds…
Figure 4. ELISpot activity of serial bleeds of volunteers in Group 1 and Group 2 with CSP peptide pools.
The ELISpot activities of each volunteer at pre-immunization, 10 d and 1, 4, 7, 10 and 11–12 m are displayed using color-coded CSP peptide pools Cp1–Cp9. The inserts show the values of the sum of each volunteer's responses at each time point and the bar indicates the geometric mean of the group. At 1 month, geomean values were higher (422 sfc/m PBMC's) in the low dose Group 1 than the high dose Group 2 (154 sfc/m PBMC's).
Figure 5. Percent of Group 1 and…
Figure 5. Percent of Group 1 and Group 2 volunteers who responded to one or more CSP or AMA1 peptide pools at each time point after immunization.
ELISpot activity was determined to be positive or negative for a given peptide pool as described in Methods. A positive volunteer is one who recognized one or more peptide pools at each of the time points 10 d and 1, 4, 7, 10 and 11–12 m after immunization. The percent positive responders are displayed at each time point for CSP or AMA1.
Figure 6. ELISpot activity of serial bleeds…
Figure 6. ELISpot activity of serial bleeds of volunteers in Group 1 and Group 2 with AMA1 peptide pools.
The ELISpot activities of each volunteer at pre-immunization, 10 d and 1, 4, 7, 10 and 11–12 m are displayed using color-coded AMA1 peptide pools Ap1–Ap12. The inserts show the values of the sum of each volunteer's responses at each time point and the bar indicates the geometric mean of the group. At 1 month, geomean values were higher (862 sfc/m PBMC's) in the low dose Group 1 than the high dose Group 2 (422 sfc/m PBMC's).
Figure 7. T-ICS CD4+ and CD8+ IFN-γ…
Figure 7. T-ICS CD4+ and CD8+ IFN-γ activity of serial bleeds of volunteers in Group 1 with CSP peptide pools.
Four Cp peptide pools that were most strongly recognized in ELISpot assays were used to determine ICS CD4+ and CD8+ IFN-γ activity. The ICS CD4+ and CD8+ T cell activities of each volunteer at 10 d and 1, 4, and 7 m (subtracting pre-immunization values) are displayed using color-coded CSP peptide pools. Scales for each phenotype have been equalized to emphasize the lower CD4+ responses. The inserts show the values of the sum of each volunteer's responses at each time point and the bar indicates the geometric mean of the group. At 1 month CD8+ responses (geomean 0.21% CD8+ T cells, std dev 0.14) were higher than CD4+ responses (geomean 0.044% CD4+ T cells, std dev 0.075).
Figure 8. T-ICS CD4+ and CD8+ IFNactivity…
Figure 8. T-ICS CD4+ and CD8+ IFNactivity of serial bleeds of each volunteer in Group 1 with AMA1 peptide pools.
Eight Ap peptide pools that were most strongly recognized in ELISpot assays were used to determine ICS CD4+ and CD8+ IFN-γ activity. The ICS CD4+ and CD8+ T cell activities of each volunteer at 10 days and 1, 4, and 7 months (subtracting pre-immunization values) are displayed using color-coded AMA1 peptide pools. Scales for each phenotype have been equalized to emphasize the lower CD4+ responses. The inserts show the values of the sum of each volunteer's responses at each time point and the bar indicates the geometric mean of the group. At 1 month, CD8+ responses (geomean 0.44% CD8+ T cells, std dev 0.58) were higher than CD4+ T-INF-γ responses (geomean 0.086% CD4+ T cells, std dev 0.17).
Figure 9. Multifunctional CD4+ and CD8+ T…
Figure 9. Multifunctional CD4+ and CD8+ T cells of serial bleeds of volunteers in Group 1 with CSP peptide pools.
A and C: Multifunctional (any two cytokines) CD4+ and CD8+ T cell activities of each volunteer at 10 d, 1, 4, and 7 m after immunization (subtracting pre-immunization values) are displayed using color-coded CSP peptide pools. At 1 month, CD4+ multi responses (geomean 0.027% CD4+ T cells, std dev 953) and CD8+ multi responses (geomean 0.06% CD8+ T cells, std dev 346) were similar. B and D: Pie charts representing the proportion of the cytokine response indicated by cytokine subsets as shown; numbers on pie charts represent percent of that subset of the total cells.
Figure 10. Multifunctional CD4+ and CD8+ T…
Figure 10. Multifunctional CD4+ and CD8+ T cells of serial bleeds of volunteers in Group 1 with AMA1 peptide pools.
A and C: Multifunctional (any two cytokines) CD4+ and CD8+ T cell activities of each volunteer at 10 d, 1, 4, and 7 m after immunization (subtracting pre-immunization values) are displayed using color-coded AMA1 peptide pools. At 1 month, CD4+ multi responses (geomean 0.072% CD4+ T cells, std dev 94) and CD8+ multi responses (geomean 0.13% CD8+ T cells, std dev 14) were similar. B and D: Pie charts representing the proportion of the cytokine response indicated by cytokine subsets as shown; numbers on pie charts represent per cent of that subset of the total cells.
Figure 11. Group 1 and Group 2…
Figure 11. Group 1 and Group 2 anti-CSP and AMA1 antibody responses by ELISA and IFA.
A and B: Box plots of the means and 25% and 75% percentiles of each group at 10 d and 1, 4, 7, 10 and 11–12 m after immunization. The first and third quartiles are the top and base of each box, and the upper and lower bars represent the high and low values respectively. C: IFA titers of each group pre-immunization and at 1 month post-immunization against sporozoites (left panel) and infected red blood cells (right panel). Bars represent geomeans.

References

    1. Clyde DF. Immunization of man against falciparum and vivax malaria by use of attenuated sporozoites. Am J Trop Med Hyg. 1975;24:397–401.
    1. Sigler CI, Leland P, Hollingdale MR. In vitro infectivity of irradiated Plasmodium berghei sporozoites to cultured hepatoma cells. Am J Trop Med Hyg. 1984;33:544–547.
    1. Nussenzweig RS, Vanderberg J, Most H, Orton C. Protective immunity produced by the injection of x-irradiated sporozoites of plasmodium berghei. Nature. 1967;216:160–162.
    1. Oliveira GA, Kumar KA, Calvo-Calle JM, Othoro C, Altszuler D, et al. Class II-restricted protective immunity induced by malaria sporozoites. Infect Immun. 2008;76:1200–1206.
    1. Doolan DL, Martinez-Alier N. Immune response to pre-erythrocytic stages of malaria parasites. Curr Mol Med. 2006;6:169–185.
    1. Jobe O, Donofrio G, Sun G, Liepinsh D, Schwenk R, et al. 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.
    1. Fandeur T, Dubois P, Gysin J, Dedet JP, da Silva LP. In vitro and in vivo studies on protective and inhibitory antibodies against Plasmodium falciparum in the Saimiri monkey. J Immunol. 1984;132:432–437.
    1. Beeson JG, Osier FH, Engwerda CR. Recent insights into humoral and cellular immune responses against malaria. Trends Parasitol. 2008;24:578–584.
    1. Marsh K, Kinyanjui S. Immune effector mechanisms in malaria. Parasite Immunol. 2006;28:51–60.
    1. Sedegah M, Brice GT, Rogers WO, Doolan DL, Charoenvit Y, et al. Persistence of protective immunity to malaria induced by DNA priming and poxvirus boosting: characterization of effector and memory CD8(+)-T-cell populations. Infect Immun. 2002;70:3493–3499.
    1. Jiang G, Charoenvit Y, Moreno A, Baraceros MF, Banania G, et al. Induction of multi-antigen multi-stage immune responses against Plasmodium falciparum in rhesus monkeys, in the absence of antigen interference, with heterologous DNA prime/poxvirus boost immunization. Malar J. 2007;6:135.
    1. Ockenhouse CF, Sun PF, Lanar DE, Wellde BT, Hall BT, et al. Phase I/IIa safety, immunogenicity, and efficacy trial of NYVAC-Pf7, a pox-vectored, multiantigen, multistage vaccine candidate for Plasmodium falciparum malaria. J Infect Dis. 1998;177:1664–1673.
    1. Bojang KA, Olodude F, Pinder M, Ofori-Anyinam O, Vigneron L, et al. Safety and immunogenicty of RTS,S/AS02A candidate malaria vaccine in Gambian children. Vaccine. 2005;23:4148–4157.
    1. Aponte JJ, Aide P, Renom M, Mandomando I, Bassat Q, et al. Safety of the RTS,S/AS02D candidate malaria vaccine in infants living in a highly endemic area of Mozambique: a double blind randomised controlled phase I/IIb trial. Lancet. 2007;370:1543–1551.
    1. Kester KE, McKinney DA, Tornieporth N, Ockenhouse CF, Heppner DG, Jr, et al. A phase I/IIa safety, immunogenicity, and efficacy bridging randomized study of a two-dose regimen of liquid and lyophilized formulations of the candidate malaria vaccine RTS,S/AS02A in malaria-naive adults. Vaccine. 2007;25:5359–5366.
    1. Sun P, Schwenk R, White K, Stoute JA, Cohen J, et al. Protective immunity induced with malaria vaccine, RTS,S, is linked to Plasmodium falciparum circumsporozoite protein-specific CD4+ and CD8+ T cells producing IFN-gamma. J Immunol. 2003;171:6961–6967.
    1. Mettens P, Dubois PM, Demoitie MA, Bayat B, Donner MN, et al. Improved T cell responses to Plasmodium falciparum circumsporozoite protein in mice and monkeys induced by a novel formulation of RTS,S vaccine antigen. Vaccine. 2008;26:1072–1082.
    1. Kester KE, Cummings JF, Ofori-Anyinam O, Ockenhouse CF, Krzych U, et al. Randomized, double-blind, phase 2a trial of falciparum malaria vaccines RTS,S/AS01B and RTS,S/AS02A in malaria-naive adults: safety, efficacy, and immunologic associates of protection. J Infect Dis. 2009;200:337–346.
    1. Li S, Locke E, Bruder J, Clarke D, Doolan DL, et al. Viral vectors for malaria vaccine development. Vaccine. 2007;25:2567–2574.
    1. Limbach KJ, Richie TL. Viral vectors in malaria vaccine development. Parasite Immunol. 2009;31:501–519.
    1. Barouch DH, Pau MG, Custers JH, Koudstaal W, Kostense S, et al. Immunogenicity of recombinant adenovirus serotype 35 vaccine in the presence of pre-existing anti-Ad5 immunity. J Immunol. 2004;172:6290–6297.
    1. Draper SJ, Moore AC, Goodman AL, Long CA, Holder AA, et al. Effective induction of high-titer antibodies by viral vector vaccines. Nat Med. 2008;14:819–821.
    1. Draper SJ, Goodman AL, Biswas S, Forbes EK, Moore AC, et al. Recombinant viral vaccines expressing merozoite surface protein-1 induce antibody- and T cell-mediated multistage protection against malaria. Cell Host Microbe. 2009;5:95–105.
    1. Rodriguez A, Goudsmit J, Companjen A, Mintardjo R, Gillissen G, et al. Impact of recombinant adenovirus serotype 35 priming versus boosting of a Plasmodium falciparum protein: characterization of T- and B-cell responses to liver-stage antigen 1. Infect Immun. 2008;76:1709–1718.
    1. Darrah PA, Patel DT, De Luca PM, Lindsay RW, Davey DF, et al. Multifunctional TH1 cells define a correlate of vaccine-mediated protection against Leishmania major. Nat Med. 2007;13:843–850.
    1. Russell WC. Adenoviruses: update on structure and function. J Gen Virol. 2009;90:1–20.
    1. Zaiss AK, Machado HB, Herschman HR. The influence of innate and pre-existing immunity on adenovirus therapy. J Cell Biochem 2009
    1. Cheng C, Gall JG, Kong WP, Sheets RL, Gomez PL, et al. Mechanism of ad5 vaccine immunity and toxicity: fiber shaft targeting of dendritic cells. PLoS Pathog. 2007;3:e25.
    1. Lore K, Adams WC, Havenga MJ, Precopio ML, Holterman L, et al. Myeloid and plasmacytoid dendritic cells are susceptible to recombinant adenovirus vectors and stimulate polyfunctional memory T cell responses. J Immunol. 2007;179:1721–1729.
    1. Stoute JA, Slaoui M, Heppner DG, Momin P, Kester KE, 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.
    1. Narum DL, Thomas AW. Differential localization of full-length and processed forms of PF83/AMA-1 an apical membrane antigen of Plasmodium falciparum merozoites. Mol Biochem Parasitol. 1994;67:59–68.
    1. Stowers AW, Kennedy MC, Keegan BP, Saul A, Long CA, et al. Vaccination of monkeys with recombinant Plasmodium falciparum apical membrane antigen 1 confers protection against blood-stage malaria. Infect Immun. 2002;70:6961–6967.
    1. Polley SD, Mwangi T, Kocken CH, Thomas AW, Dutta S, et al. Human antibodies to recombinant protein constructs of Plasmodium falciparum Apical Membrane Antigen 1 (AMA1) and their associations with protection from malaria. Vaccine. 2004;23:718–728.
    1. Silvie O, Franetich JF, Charrin S, Mueller MS, Siau A, et al. A role for apical membrane antigen 1 during invasion of hepatocytes by Plasmodium falciparum sporozoites. J Biol Chem. 2004;279:9490–9496.
    1. Peduzzi E, Westerfeld N, Zurbriggen R, Pluschke G, Daubenberger CA. Contribution of influenza immunity and virosomal-formulated synthetic peptide to cellular immune responses in a phase I subunit malaria vaccine trial. Clin Immunol. 2008;127:188–197.
    1. Okitsu SL, Silvie O, Westerfeld N, Curcic M, Kammer AR, et al. A virosomal malaria peptide vaccine elicits a long-lasting sporozoite-inhibitory antibody response in a phase 1a clinical trial. PLoS One. 2007;2:e1278.
    1. Sprangers MC, Lakhai W, Koudstaal W, Verhoeven M, Koel BF, et al. Quantifying adenovirus-neutralizing antibodies by luciferase transgene detection: addressing preexisting immunity to vaccine and gene therapy vectors. J Clin Microbiol. 2003;41:5046–5052.
    1. Bruder JT, Angov E, Limbach KJ, Richie TL. Molecular vaccines for malaria. Hum Vaccin. 2010;6:54–77.
    1. Brough DE, Lizonova A, Hsu C, Kulesa VA, Kovesdi I. A gene transfer vector-cell line system for complete functional complementation of adenovirus early regions E1 and E4. J Virol. 1996;70:6497–6501.
    1. Reyes-Sandoval A, Sridhar S, Berthoud T, Moore AC, Harty JT, et al. Single-dose immunogenicity and protective efficacy of simian adenoviral vectors against Plasmodium berghei. Eur J Immunol. 2008;38:732–741.
    1. Harro CD, Robertson MN, Lally MA, O'Neill LD, Edupuganti S, et al. Safety and immunogenicity of adenovirus-vectored near-consensus HIV type 1 clade B gag vaccines in healthy adults. AIDS Res Hum Retroviruses. 2009;25:103–114.
    1. Doolan DL, Hoffman SL, Southwood S, Wentworth PA, Sidney J, et al. Degenerate cytotoxic T cell epitopes from P. falciparum restricted by multiple HLA-A and HLA-B supertype alleles. Immunity. 1997;7:97–112.
    1. Blum-Tirouvanziam U, Servis C, Habluetzel A, Valmori D, Men Y, et al. Localization of HLA-A2.1-restricted T cell epitopes in the circumsporozoite protein of Plasmodium falciparum. J Immunol. 1995;154:3922–3931.
    1. Good MF, Pombo D, Quakyi IA, Riley EM, Houghten RA, et al. Human T-cell recognition of the circumsporozoite protein of Plasmodium falciparum: immunodominant T-cell domains map to the polymorphic regions of the molecule. Proc Natl Acad Sci U S A. 1988;85:1199–1203.
    1. Zevering Y, Houghten RA, Frazer IH, Good MF. Major population differences in T cell response to a malaria sporozoite vaccine candidate. Int Immunol. 1990;2:945–955.
    1. Lal AA, Hughes MA, Oliveira DA, Nelson C, Bloland PB, et al. Identification of T-cell determinants in natural immune responses to the Plasmodium falciparum apical membrane antigen (AMA-1) in an adult population exposed to malaria. Infect Immun. 1996;64:1054–1059.
    1. Wang R, Epstein J, Baraceros FM, Gorak EJ, Charoenvit Y, 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;98:10817–10822.
    1. Stewart VA, McGrath SM, Dubois PM, Pau MG, Mettens P, et al. Priming with an adenovirus 35-circumsporozoite protein (CS) vaccine followed by RTS,S/AS01B boosting significantly improves immunogenicity to Plasmodium falciparum CS compared to that with either malaria vaccine alone. Infect Immun. 2007;75:2283–2290.
    1. Spring MD, Cummings JF, Ockenhouse CF, Dutta S, Reidler R, et al. Phase 1/2a study of the malaria vaccine candidate apical membrane antigen-1 (AMA-1) administered in adjuvant system AS01B or AS02A. PLoS One. 2009;4:e5254.
    1. Charoenvit Y, Mellouk S, Cole C, Bechara R, Leef MF, et al. Monoclonal, but not polyclonal, antibodies protect against Plasmodium yoelii sporozoites. J Immunol. 1991;146:1020–1025.
    1. Bergmann-Leitner ES, Duncan EH, Mullen GE, Burge JR, Khan F, et al. Critical evaluation of different methods for measuring the functional activity of antibodies against malaria blood stage antigens. Am J Trop Med Hyg. 2006;75:437–442.
    1. Neter J. Applied Linear Statistical Methods. WCB McGraw-Hill, New York; 1996.
    1. Moorthy VS, Imoukhuede EB, Keating S, Pinder M, Webster D, et al. Phase 1 evaluation of 3 highly immunogenic prime-boost regimens, including a 12-month reboosting vaccination, for malaria vaccination in Gambian men. J Infect Dis. 2004;189:2213–2219.
    1. Vuola JM, Keating S, Webster DP, Berthoud T, Dunachie S, et al. Differential immunogenicity of various heterologous prime-boost vaccine regimens using DNA and viral vectors in healthy volunteers. J Immunol. 2005;174:449–455.
    1. Harro C, Sun X, Stek JE, Leavitt RY, Mehrotra DV, et al. Safety and immunogenicity of the Merck adenovirus serotype 5 (MRKAd5) and MRKAd6 human immunodeficiency virus type 1 trigene vaccines alone and in combination in healthy adults. Clin Vaccine Immunol. 2009;16:1285–1292.
    1. Priddy FH, Brown D, Kublin J, Monahan K, Wright DP, et al. Safety and immunogenicity of a replication-incompetent adenovirus type 5 HIV-1 clade B gag/pol/nef vaccine in healthy adults. Clin Infect Dis. 2008;46:1769–1781.
    1. Seder RA, Darrah PA, Roederer M. T-cell quality in memory and protection: implications for vaccine design. Nat Rev Immunol. 2008;8:247–258.
    1. Kaufhold RM, Field JA, Caulfield MJ, Wang S, Joseph H, et al. Memory T-cell response to rotavirus detected with a gamma interferon enzyme-linked immunospot assay. J Virol. 2005;79:5684–5694.
    1. Minigo G, Woodberry T, Piera KA, Salwati E, Tjitra E, et al. Parasite-dependent expansion of TNF receptor II-positive regulatory T cells with enhanced suppressive activity in adults with severe malaria. PLoS Pathog. 2009;5:e1000402.
    1. Doolan DL, Southwood S, Chesnut R, Appella E, Gomez E, et al. HLA-DR-promiscuous T cell epitopes from Plasmodium falciparum pre-erythrocytic-stage antigens restricted by multiple HLA class II alleles. J Immunol. 2000;165:1123–1137.
    1. Sedegah M, Kim Y, Peters B, McGrath S, Ganeshan H, et al. Identification and localization of minimal MHC-restricted CD8+ T cell epitopes within the Plasmodium falciparum AMA1 protein. Malar J. 9:241.
    1. Bongfen SE, Ntsama PM, Offner S, Smith T, Felger I, et al. The N-terminal domain of Plasmodium falciparum circumsporozoite protein represents a target of protective immunity. Vaccine. 2009;27:328–335.
    1. Wang R, Doolan DL, Le TP, Hedstrom RC, Coonan KM, et al. Induction of antigen-specific cytotoxic T lymphocytes in humans by a malaria DNA vaccine. Science. 1998;282:476–480.
    1. Nardin EH, Calvo-Calle JM, Oliveira GA, Nussenzweig RS, Schneider M, et al. A totally synthetic polyoxime malaria vaccine containing Plasmodium falciparum B cell and universal T cell epitopes elicits immune responses in volunteers of diverse HLA types. J Immunol. 2001;166:481–489.
    1. Lockyer MJ, Marsh K, Newbold CI. Wild isolates of Plasmodium falciparum show extensive polymorphism in T cell epitopes of the circumsporozoite protein. Mol Biochem Parasitol. 1989;37:275–280.
    1. Stoute JA, Kester KE, Krzych U, Wellde BT, Hall T, et al. Long-term efficacy and immune responses following immunization with the RTS,S malaria vaccine. J Infect Dis. 1998;178:1139–1144.
    1. Udhayakumar V, Kariuki S, Kolczack M, Girma M, Roberts JM, et al. Longitudinal study of natural immune responses to the Plasmodium falciparum apical membrane antigen (AMA-1) in a holoendemic region of malaria in western Kenya: Asembo Bay Cohort Project VIII. Am J Trop Med Hyg. 2001;65:100–107.
    1. Huaman MC, Mullen GE, Long CA, Mahanty S. Plasmodium falciparum apical membrane antigen 1 vaccine elicits multifunctional CD4 cytokine-producing and memory T cells. Vaccine. 2009;27:5239–5246.
    1. Reyes-Sandoval A, Berthoud T, Alder N, Siani L, Gilbert SC, 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. 78:145–153.
    1. Shott JP, McGrath SM, Pau MG, Custers JH, Ophorst O, et al. Adenovirus 5 and 35 vectors expressing Plasmodium falciparum circumsporozoite surface protein elicit potent antigen-specific cellular IFN-gamma and antibody responses in mice. Vaccine 2008
    1. Liu J, Ewald BA, Lynch DM, Denholtz M, Abbink P, et al. Magnitude and phenotype of cellular immune responses elicited by recombinant adenovirus vectors and heterologous prime-boost regimens in rhesus monkeys. J Virol. 2008;82:4844–4852.
    1. Bruna-Romero O, Rocha CD, Tsuji M, Gazzinelli RT. Enhanced protective immunity against malaria by vaccination with a recombinant adenovirus encoding the circumsporozoite protein of Plasmodium lacking the GPI-anchoring motif. Vaccine. 2004;22:3575–3584.
    1. Pierce MA, Ellis RD, Martin LB, Malkin E, Tierney E, et al. Phase 1 safety and immunogenicity trial of the Plasmodium falciparum blood-stage malaria vaccine AMA1-C1/ISA 720 in Australian adults. Vaccine. 28:2236–2242.
    1. Malkin EM, Diemert DJ, McArthur JH, Perreault JR, Miles AP, et al. Phase 1 clinical trial of apical membrane antigen 1: an asexual blood-stage vaccine for Plasmodium falciparum malaria. Infect Immun. 2005;73:3677–3685.
    1. Bruder JT, Stefaniak ME, Patterson NB, Chen P, Konovalova S, et al. Adenovectors induce functional antibodies capable of potent inhibition of blood stage malaria parasite growth. Vaccine. 2010;28:3201–3210.
    1. Koup RA, Roederer M, Lamoreaux L, Fischer J, Novik 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. Kumar KA, Sano G, Boscardin S, Nussenzweig RS, Nussenzweig MC, et al. The circumsporozoite protein is an immunodominant protective antigen in irradiated sporozoites. Nature. 2006;444:937–940.
    1. Hoffman SL, Goh LM, Luke TC, Schneider I, Le TP, et al. Protection of humans against malaria by immunization with radiation-attenuated Plasmodium falciparum sporozoites. J Infect Dis. 2002;185:1155–1164.
    1. Doolan DL, Southwood S, Freilich DA, Sidney J, Graber NL, et al. Identification of Plasmodium falciparum antigens by antigenic analysis of genomic and proteomic data. Proc Natl Acad Sci U S A. 2003;100:9952–9957.
    1. Egan JE, Hoffman SL, Haynes JD, Sadoff JC, Schneider I, et al. Humoral immune responses in volunteers immunized with irradiated Plasmodium falciparum sporozoites. Am J Trop Med Hyg. 1993;49:166–173.
    1. Hollingdale MR, Appiah A, Leland P, do Rosario VE, Mazier D, et al. Activity of human volunteer sera to candidate Plasmodium falciparum circumsporozoite protein vaccines in the inhibition of sporozoite invasion assay of human hepatoma cells and hepatocytes. Trans R Soc Trop Med Hyg. 1990;84:325–329.
    1. Lindsay RW, Darrah PA, Quinn KM, Wille-Reece U, Mattei LM, et al. CD8+ T cell responses following replication-defective adenovirus serotype 5 immunization are dependent on CD11c+ dendritic cells but show redundancy in their requirement of TLR and nucleotide-binding oligomerization domain-like receptor signaling. J Immunol. 185:1513–1521.
    1. Geiben-Lynn R, Greenland JR, Frimpong-Boateng K, Letvin NL. Kinetics of recombinant adenovirus type 5, vaccinia virus, modified vaccinia ankara virus, and DNA antigen expression in vivo and the induction of memory T-lymphocyte responses. Clin Vaccine Immunol. 2008;15:691–696.

Source: PubMed

3
Tilaa