Sterile immunity to malaria after DNA prime/adenovirus boost immunization is associated with effector memory CD8+T cells targeting AMA1 class I epitopes

Martha Sedegah, Michael R Hollingdale, Fouzia Farooq, Harini Ganeshan, Maria Belmonte, Yohan Kim, Bjoern Peters, Alessandro Sette, Jun Huang, Shannon McGrath, Esteban Abot, Keith Limbach, Meng Shi, Lorraine Soisson, Carter Diggs, Ilin Chuang, Cindy Tamminga, Judith E Epstein, Eileen Villasante, Thomas L Richie, Martha Sedegah, Michael R Hollingdale, Fouzia Farooq, Harini Ganeshan, Maria Belmonte, Yohan Kim, Bjoern Peters, Alessandro Sette, Jun Huang, Shannon McGrath, Esteban Abot, Keith Limbach, Meng Shi, Lorraine Soisson, Carter Diggs, Ilin Chuang, Cindy Tamminga, Judith E Epstein, Eileen Villasante, Thomas L Richie

Abstract

Background: Fifteen volunteers were immunized with three doses of plasmid DNA encoding P. falciparum circumsporozoite protein (CSP) and apical membrane antigen-1 (AMA1) and boosted with human adenovirus-5 (Ad) expressing the same antigens (DNA/Ad). Four volunteers (27%) demonstrated sterile immunity to controlled human malaria infection and, overall, protection was statistically significantly associated with ELISpot and CD8+ T cell IFN-γ activities to AMA1 but not CSP. DNA priming was required for protection, as 18 additional subjects immunized with Ad alone (AdCA) did not develop sterile protection.

Methodology/principal findings: We sought to identify correlates of protection, recognizing that DNA-priming may induce different responses than AdCA alone. Among protected volunteers, two and three had higher ELISpot and CD8+ T cell IFN-γ responses to CSP and AMA1, respectively, than non-protected volunteers. Unexpectedly, non-protected volunteers in the AdCA trial showed ELISpot and CD8+ T cell IFN-γ responses to AMA1 equal to or higher than the protected volunteers. T cell functionality assessed by intracellular cytokine staining for IFN-γ, TNF-α and IL-2 likewise did not distinguish protected from non-protected volunteers across both trials. However, three of the four protected volunteers showed higher effector to central memory CD8+ T cell ratios to AMA1, and one of these to CSP, than non-protected volunteers for both antigens. These responses were focused on discrete regions of CSP and AMA1. Class I epitopes restricted by A*03 or B*58 supertypes within these regions of AMA1 strongly recalled responses in three of four protected volunteers. We hypothesize that vaccine-induced effector memory CD8+ T cells recognizing a single class I epitope can confer sterile immunity to P. falciparum in humans.

Conclusions/significance: We suggest that better understanding of which epitopes within malaria antigens can confer sterile immunity and design of vaccine approaches that elicit responses to these epitopes will increase the potency of next generation gene-based vaccines.

Trial registration: ClinicalTrials.gov NCT00392015 NCT00870987.

Conflict of interest statement

Competing Interests: CD and LS from USAID (funders) played a role in study design. This does not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Figure 1. DNA/Ad Trial design.
Figure 1. DNA/Ad Trial design.
DNA/Ad Trial: Healthy, malaria-naïve adult research subjects received three doses of two DNA plasmids (mixture of 1 mg CSP and 1 mg AMA1, 2 mg total dose, in 2 ml phosphate buffered saline) in two divided doses of 1 ml into each deltoid muscle by needleless jet injection, followed by one dose of two adenovectors (mixture of 1×1010 particle units CSP and 1×1010 particle units of AMA1, 2×1010 particle units total dose, in 1 ml final formulation buffer) into one deltoid muscle by needle and syringe. Both Ad5 seropositive and seronegative study subjects were enrolled. AdCA Trial: Malaria-naïve adult research subjects received one dose of two adenovectors (formulated identically as in DNA/Ad) into one deltoid muscle by needle and syringe. Only Ad5 seronegative study subjects (neutralizing antibody titer <500, NVITAL assay) were enrolled. CHMI  =  controlled human malaria infection, administered via the bites of five laboratory-reared P. falciparum-infected Anopheles stephensi mosquitoes.
Figure 2. DNA/Ad ELISpot IFN-γ activities to…
Figure 2. DNA/Ad ELISpot IFN-γ activities to CSP and AMA1 peptide pools.
Panels A and C: ELISpot activities (sfc/m) of each volunteer to CSP or AMA1 peptide pools are shown as color coded stacked bars at pre-immunization (1), 28 days after DNA immunization (2) and 22/23 days after the Ad boost (3). *Positive activities. Panels B and D: ELISpot activities were summed and protected subjects shown as color-coded dots. Horizontal bars represent geometric mean activities of non-protected volunteers. The geometric means of summed activities of non-protected volunteers and the activities of each protected volunteer are shown in Table 1. Panel A: CSP: All volunteers were negative after DNA immunization, but four volunteers were positive after Ad boost including protected v11 (408 sfc/m) and v18 (398 sfc/m) that were above non-protected volunteers, whereas protected v06 and v10 were negative. Most activity of v11 and v18 was directed to single CSP peptide pools. Panel B: CSP: Geometric means of non-protected volunteers remained similar after DNA immunization and Ad boost when v11 and v18 were higher than all other volunteers. Panel C: AMA1: 11 volunteers were positive after DNA immunization (*gray), and 11 were positive after Ad boost (*black), including protected v10 (810 sfc/m), v11 (1046 sfc/m) and v18 (1270 sfc/m) that were above non-protected volunteers, whereas protected v06 was within the range of non-protected volunteers. Most activity of v10, v11 and v18 was directed to single AMA1 peptide pools. Panel D: AMA1: Geometric means of non-protected volunteers were higher Ad boost when v10, v11 and v18 higher than all volunteers.
Figure 3. DNA/Ad CD8+ T cell IFN-γ…
Figure 3. DNA/Ad CD8+ T cell IFN-γ activities to CSP and AMA1 peptide pools.
Panels A and C: CD8+ T cell IFN-γ activities of each volunteer to CSP or AMA1 peptide pools are shown as color coded stacked bars at pre-immunization (1), 28 days after DNA immunization (2) and 22/23 days after the Ad boost (3). *Positive activities (gray: post-DNA: black: post-Ad). Panels B and D: CD8+ T cell IFN-γ activities were summed and are shown as color-coded dots. Horizontal bars represent geometric mean activities of non-protected volunteers. The geometric means of summed activities of non-protected volunteers and the activities of each protected volunteer are shown in Table 1. Panel A: CSP: Five non-protected volunteers were positive after DNA immunization, but four volunteers were positive after Ad boost including protected v11 (0.21%) and v18 (0.10%) that were above non-protected volunteers, except v15 that had highest activity (0.29%), whereas protected v06 and v10 were negative. Most activity of v11, v18 and v15 was directed to single CSP peptide pools. Panel B: CSP: Geometric means of non-protected volunteers remained similar after DNA immunization and Ad boost when v11 and v18 were higher than all other volunteers, except v15 (arrow). Panel C: AMA1: Four non-protected volunteers were positive after DNA immunization, and 11 were positive after Ad boost, including protected v10 (0.22%), v11 (0.98%) and v18 (0.52%) that were above non-protected volunteers, whereas protected v06 negative. Most activity of v10, v11 and v18 was directed to single AMA1 peptide pools. Panel D: AMA1: Geometric means of non-protected volunteers rose after Ad boost compared to after DNA immunization, and v10, v11 and v18 higher than all volunteers.
Figure 4. DNA/Ad CD8+ T cell memory…
Figure 4. DNA/Ad CD8+ T cell memory IFN-γ activities to CSP and AMA1 peptide pools.
Panels A and C: CD8+ T cell IFN-γ activities of each volunteer to CSP or AMA1 peptide pools 22/23 days after the Ad boost are shown as color coded stacked bars, differentiating memory naïve (N), central (C), effector (E) and terminally differentiated (T) cells (per cent of CD8+ T cells) to CSP and AMA1 peptides as color-coded bars. Non-protected and protected volunteers are grouped. *Positive activities (defined in Methods). Panels B and D: CD8+ T cell IFN-γ memory activities were summed and are shown as color-coded dots. Horizontal bars represent geometric mean activities of non-protected volunteers. The geometric means of summed activities of non-protected volunteers and the activities of each protected volunteer are shown in Table 1. Panel A: CSP: among non-protected volunteers, only one (v15) had positive CM and TD activities and none had positive EM activities. One protected volunteer (v11) had positive CM, EM and TD activities whereas protected v18 had only positive CM activity. Positive CM, EM and TD activities were predominantly directed to single CSP peptide pools. Panel B: the geometric mean of CM and EM activities of non-protected volunteers were lower than protected v11 and v18, except CM activity of non-protected v15 (arrow). Panel C: AMA1: five non-protected volunteers only had positive CM activities, whereas protected v10 had positive CM and EM, and v11 and v18 had positive CM, EM and TD activities that were mostly directed to single AMA1 peptide pools. Panel D: AMA1: the geometric mean of CM activities of non-protected volunteers was similar to v10 but lower than v11 and v18; however, protected v10, v11 and v18 were above the geometric means of EM and TD activities of non-protected volunteers.
Figure 5. DNA/Ad and AdCA CD8+ T…
Figure 5. DNA/Ad and AdCA CD8+ T cell antigen-specific activities are predominantly monofunctional with lower polyfunctional responses.
Monofunctional and polyfunctional CD8+ T cell activities to CSP and AMA1 after DNA/Ad (Panels A and B) and AdCA (Panels C and D) immunization are shown as color-coded filled circles that represent the percent of CD8+ T cells containing cytokine(s). Black horizontal bar denotes highest activities in DNA/Ad compared with AdCA activities. Activities to individual CSP and AMA1 peptide pools are shown in Figures S1 and S2. Panel A: DNA/Ad CSP: IFN-γ monofunctional activities of protected v11 and v18 were higher (v11, 0.14%; v18, 0.1%) than the two positive non-protected volunteers (v03, 0.07%; v15, 0.09%). Non-protected v15 developed the highest IFN-γ/IL2 polyfunctional (0.20%) and IL2 monofunctional (0.5%) activities. Protected v11 developed positive IFN-γ/IL2 polyfunctional (0.07%) and IL2 monofunctional (0.06%) activities. Panel B: DNA/Ad AMA1: IFN-γ monofunctional activities of protected v10, v11 and v18 (v10, 0.16%; v11, 0.58%, v18, 0.49%) were higher than the eight non-protected volunteers (range 0.05–0.14%). Non-protected v17 and protected v10 also developed lower TNF-α monofunctional (0.07%, 0.09%) activity; v11 also developed IFN-γ/TNF-α polyfunctional (0.31%) activity and lower IFN-γ/IL2 polyfunctional (0.04%), and IFN-γ/IL2/TNF-α polyfunctional (0.05%) activities. Panel C: AdCA CSP: 11 volunteers developed IFN-γ monofunctional activities (0.05%–0.48%), of whom one volunteer developed IFN-γ/IL2 polyfunctional, three developed IL2 monofunctional and two additional volunteers also developed IL2 monofunctional activities. Four volunteers had higher IFN-γ and two volunteers had higher IL2 monofunctional activities than protected volunteers in DNA/Ad. Panel D: AdCA AMA1: all 16 volunteers developed positive IFN-γ monofunctional – boxed volunteers are above scale (1.60% and 1.79% respectively), of whom nine developed lower IFN-γ/IL2 polyfunctional or IFN-γ/TNF-α polyfunctional activities; three volunteers developed IL2 monofunctional and TNF-α monofunctional activities. Four volunteers had higher IFN-γ monofunctional and eight volunteers had higher IFN-γ/IL2 polyfunctional activities than protected volunteers in the DNA/Ad trial.
Figure 6. Ratios of effector to central…
Figure 6. Ratios of effector to central memory in DNA/Ad and AdCA trials for CSP and AMA1 (based on summed and pool-specific responses).
All volunteers who had positive summed and pool-specific CD8+ IFN-γ memory central (CM) and effector (EM) activities after Ad boost for CSP or AMA1 were plotted as the ratio of EM:CM vs. EM activity. In addition, some volunteers who had positive CM but negative EM activities were included. Volunteers with negative CM and EM activities were not included. Panel A. CSP summed response: protected v11 and v18 had a higher EM:CM ratio than all other volunteers in the DNA/Ad and AdCA trials, except v135 in the AdCA trial (*). EM activity of v18 did not meet the positivity definition. Panel B. CSP pool-specific response: protected v11 and v18 had a higher EM:CM ratios against Cp9 (v11) and Cp6 (v18) respectively than all other volunteers with positive EM and/or CM activities against any individual pools in the DNA/Ad and AdCA trials, except v135 in the AdCA trial. EM activity of v18 did not meet the positivity definition. Panel C. AMA1 summed response: protected v10, v11, and v18 had a higher EM:CM ratio than all other volunteers with positive EM and/or CM activities in the DNA/Ad and AdCA trials. Panel D. AMA1 pool specific response: protected v10, v11 and v18 had a higher EM:CM ratios against Ap8 (v10 and v18) and Ap10 (v11) compared to all other volunteers in the DNA/Ad and AdCA trials.
Figure 7. DNA/Ad and AdCA: Comparison of…
Figure 7. DNA/Ad and AdCA: Comparison of positive T cell activities and percent of total activities to CSP peptide pools.
ELISpot, CD8+ T cell IFN-γ, and CD8+ T cell EM IFN-γ activities to individual CSP peptide pools (Cp6 and Cp9) were calculated as per cent of total activities of summed responses to all CSP peptide pools. All volunteers positive with Cp6 or Cp9 were selected. Panels A, B, C: Cp6: protected v18 had lower activities than some AdCA volunteers, but highest per cent activities; v194 (delay to patency) had higher per cent ELISpot activity than other AdCA volunteers, but lower than v18. None of the DNA/Ad or AdCA volunteers had positive EM activity to Cp6. Panels D, E, F: Cp9: protected v11 had highest per cent ELISpot activity to Cp9, but CD8+ T cell IFN-γ and EM activities were lower than some AdCA volunteers.
Figure 8. DNA/Ad and AdCA: Comparison of…
Figure 8. DNA/Ad and AdCA: Comparison of positive T cell activities and per cent of total activities to AMA1 peptide pools.
ELISpot IFN-γ, CD8+ T cell IFN-γ, and CD8+ T cell EM IFN-γ activities to individual AMA1 peptide pools (Ap8 and Ap10) were calculated as per cent of total activities of summed responses to all AMA1 peptide pools. All volunteers positive with Ap8 or Ap10 were selected. Panels A, B, C: Ap8: protected v10 and v18 had lower activities than one or more AdCA volunteers, but highest per cent activities; v194 (delay to patency, blue arrows) had higher per cent ELISpot and CD8+ T cell IFN-γ activities than other AdCA volunteers, but no CD8+ T cell IFN-γ EM activity. Panels D, E, F: Ap10: protected v11 had highest per cent ELISpot, CD8+ T cell IFN-γ and CD8+ T cell IFN-γ EM activities to Ap10.

References

    1. Murray CJ, Rosenfeld LC, Lim SS, Andrews KG, Foreman KJ, et al. (2012) Global malaria mortality between 1980 and 2010: a systematic analysis. Lancet 379: 413–431.
    1. Das P, Horton R (2010) Malaria elimination: worthy, challenging, and just possible. Lancet 376: 1515–1517.
    1. Weiss WR, Good MF, Hollingdale MR, Miller LH, Berzofsky JA (1989) Genetic control of immunity to Plasmodium yoelii sporozoites. J Immunol 143: 4263–4266.
    1. Hoffman SL, Weiss W, Mellouk S, Sedegah M (1990) Irradiated sporozoite vaccine induces cytotoxic T lymphocytes that recognize malaria antigens on the surface of infected hepatocytes. Immunol Lett 25: 33–38.
    1. Weiss WR, Jiang CG (2012) Protective CD8+ T lymphocytes in primates immunized with malaria sporozoites. PLoS One 7: e31247.
    1. Overstreet MG, Cockburn IA, Chen YC, Zavala F (2008) Protective CD8 T cells against Plasmodium liver stages: immunobiology of an ‘unnatural’ immune response. Immunol Rev 225: 272–283.
    1. Krzych U, Dalai S, Zarling S, Pichugin A (2012) Memory CD8 T cells specific for plasmodia liver-stage antigens maintain protracted protection against malaria. Front Immunol 3: 370.
    1. Doolan DL, Martinez-Alier N (2006) Immune response to pre-erythrocytic stages of malaria parasites. Curr Mol Med 6: 169–185.
    1. Butler NS, Schmidt NW, Vaughan AM, Aly AS, Kappe SH, et al. (2011) Superior antimalarial immunity after vaccination with late liver stage-arresting genetically attenuated parasites. Cell Host Microbe 9: 451–462.
    1. Gilbert SC, Schneider J, Hannan CM, Hu JT, Plebanski M, et al. (2002) Enhanced CD8 T cell immunogenicity and protective efficacy in a mouse malaria model using a recombinant adenoviral vaccine in heterologous prime-boost immunisation regimes. Vaccine 20: 1039–1045.
    1. Sedegah M, Hedstrom R, Hobart P, Hoffman SL (1994) Protection against malaria by immunization with plasmid DNA encoding circumsporozoite protein. Proc Natl Acad Sci U S A 91: 9866–9870.
    1. Jiang G, Shi M, Conteh S, Richie N, Banania G, et al. (2009) Sterile protection against Plasmodium knowlesi in rhesus monkeys from a malaria vaccine: comparison of heterologous prime boost strategies. PLoS One 4: e6559.
    1. Moorthy VS, Imoukhuede EB, Milligan P, Bojang K, Keating S, et al. (2004) A randomised, double-blind, controlled vaccine efficacy trial of DNA/MVA ME-TRAP against malaria infection in Gambian adults. PLoS Med 1: e33.
    1. Bejon P, Mwacharo J, Kai OK, Todryk S, Keating S, et al. (2006) Immunogenicity of the candidate malaria vaccines FP9 and modified vaccinia virus Ankara encoding the pre-erythrocytic antigen ME-TRAP in 1–6 year old children in a malaria endemic area. Vaccine 24: 4709–4715.
    1. Porter DW, Thompson FM, Berthoud TK, Hutchings CL, Andrews L, et al. (2011) A human Phase I/IIa malaria challenge trial of a polyprotein malaria vaccine. Vaccine 29: 7514–7522.
    1. Sheehy SH, Duncan CJ, Elias SC, Biswas S, Collins KA, et al. (2012) 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 7: e31208.
    1. Ogwang C, Afolabi M, Kimani D, Jagne YJ, Sheehy SH, et al. (2013) Safety and immunogenicity of heterologous prime-boost immunisation with Plasmodium falciparum malaria candidate vaccines, ChAd63 ME-TRAP and MVA ME-TRAP, in healthy Gambian and Kenyan adults. PLoS One 8: e57726.
    1. Webster DP, Dunachie S, Vuola JM, Berthoud T, Keating S, et al. (2005) 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 102: 4836–4841.
    1. Chuang I, Sedegah M, Cicatelli S, Spring M, Polhemus M, et al. (2013) DNA prime/adenovirus boost malaria vaccine encoding P. falciparum CSP and AMA1 induces sterile protection associated with cell-mediated immunity PLoS One. 8: 1371.
    1. Ewer KJ, O′Hara GA, Duncan CJ, Collins KA, Sheehy SH, et al. (2013) Protective CD8+ T-cell immunity to human malaria induced by chimpanzee adenovirus-MVA immunisation. Nat Commun 4: 2836.
    1. Silvie O, Franetich JF, Charrin S, Mueller MS, Siau A, et al. (2004) A role for apical membrane antigen 1 during invasion of hepatocytes by Plasmodium falciparum sporozoites. J Biol Chem 279: 9490–9496.
    1. Nussenzweig V, Nussenzweig RS (1985) Circumsporozoite proteins of malaria parasites. Cell 42: 401–403.
    1. Lumsden JM, Schwenk RJ, Rein LE, Moris P, Janssens M, et al. (2011) Protective immunity induced with the RTS, S/AS vaccine is associated with IL-2 and TNF-alpha producing effector and central memory CD4 T cells. PLoS One 6: e20775.
    1. Polley SD, Mwangi T, Kocken CH, Thomas AW, Dutta S, et al. (2004) Human antibodies to recombinant protein constructs of Plasmodium falciparum Apical Membrane Antigen 1 (AMA1) and their associations with protection from malaria. Vaccine 23: 718–728.
    1. Sedegah M, Tamminga C, McGrath S, House B, Ganeshan H, et al. (2011) Adenovirus 5-vectored P. falciparum Vaccine Expressing CSP and AMA1. Part A: Safety and Immunogenicity in Seronegative Adults. PLoS One 6: e24586.
    1. Tamminga C, Sedegah M, Maiolatesi S, Fedders C, Reyes S, et al. (2013) 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. Hum Vaccin Immunother 9: 2165–2177.
    1. Seder RA, Darrah PA, Roederer M (2008) T-cell quality in memory and protection: implications for vaccine design. Nat Rev Immunol 8: 247–258.
    1. Epstein JE, Tewari K, Lyke KE, Sim BK, Billingsley PF, et al... (2011) Live Attenuated Malaria Vaccine Designed to Protect through Hepatic CD8+ T Cell Immunity. Science.
    1. Seder RA, Chang LJ, Enama ME, Zephir KL, Sarwar UN, et al. (2013) Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science 341: 1359–1365.
    1. Kester KE, McKinney DA, Tornieporth N, Ockenhouse CF, Heppner DG Jr, et al. (2007) 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 25: 5359–5366.
    1. Wang R, Epstein J, Baraceros FM, Gorak EJ, Charoenvit Y, et al. (2001) Induction of CD4(+) T cell-dependent CD8(+) type 1 responses in humans by a malaria DNA vaccine. Proc Natl Acad Sci U S A 98: 10817–10822.
    1. Tamminga C, Sedegah M, Regis D, Chuang I, Epstein JE, et al. (2011) Adenovirus-5-vectored P. falciparum vaccine expressing CSP and AMA1. Part B: safety, immunogenicity and protective efficacy of the CSP component. PLoS One 6: e25868.
    1. Sedegah M, Kim D, Ganeshan H, Huang J, Belmonte M, et al. (2013) Identification of minimal human MHC-restricted CD8+ T-cell epitopes within the Plasmodium falciparum circumsporozoite protein (CSP). Malar J 12: 185.
    1. Hamann D, Baars PA, Rep MH, Hooibrink B, Kerkhof-Garde SR, et al. (1997) Phenotypic and functional separation of memory and effector human CD8+ T cells. J Exp Med 186: 1407–1418.
    1. Vasconcelos JR, Dominguez MR, Araujo AF, Ersching J, Tararam CA, et al. (2012) Relevance of long-lived CD8(+) T effector memory cells for protective immunity elicited by heterologous prime-boost vaccination. Front Immunol 3: 358.
    1. Harro CD, Robertson MN, Lally MA, O′Neill LD, Edupuganti S, et al. (2009) Safety and immunogenicity of adenovirus-vectored near-consensus HIV type 1 clade B gag vaccines in healthy adults. AIDS Res Hum Retroviruses 25: 103–114.
    1. Reyes-Sandoval A, Berthoud T, Alder N, Siani L, Gilbert SC, et al. (2010) 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. Wherry EJ (2009) T cell exhaustion. Nat Immunol 12: 492–499.
    1. Lundegaard C, Lund O, Nielsen M (2008) Accurate approximation method for prediction of class I MHC affinities for peptides of length 8, 10 and 11 using prediction tools trained on 9mers. Bioinformatics 24: 1397–1398.
    1. Rammensee H, Bachmann J, Emmerich NP, Bachor OA, Stevanovic S (1999) SYFPEITHI: database for MHC ligands and peptide motifs. Immunogenetics 50: 213–219.
    1. Sidney J, Peters B, Frahm N, Brander C, Sette A (2008) HLA class I supertypes: a revised and updated classification. BMC Immunol 9: 1.
    1. Dunachie SJ, Walther M, Epstein JE, Keating S, Berthoud T, et al. (2006) A DNA prime-modified vaccinia virus ankara boost vaccine encoding thrombospondin-related adhesion protein but not circumsporozoite protein partially protects healthy malaria-naive adults against Plasmodium falciparum sporozoite challenge. Infect Immun 74: 5933–5942.
    1. Priddy FH, Brown D, Kublin J, Monahan K, Wright DP, et al. (2008) Safety and immunogenicity of a replication-incompetent adenovirus type 5 HIV-1 clade B gag/pol/nef vaccine in healthy adults. Clin Infect Dis 46: 1769–1781.
    1. Miao J, Li X, Liu Z, Xue C, Bujard H, et al. (2006) Immune responses in mice induced by prime-boost schemes of the Plasmodium falciparum apical membrane antigen 1 (PfAMA1)-based DNA, protein and recombinant modified vaccinia Ankara vaccines. Vaccine 24: 6187–6198.
    1. Bouillet LE, Dias MO, Dorigo NA, Moura AD, Russell B, et al. (2011) Long-term humoral and cellular immune responses elicited by a heterologous Plasmodium vivax apical membrane antigen 1 protein prime/adenovirus boost immunization protocol. Infect Immun 79: 3642–3652.
    1. Steffensen MA, Holst PJ, Steengaard SS, Jensen BA, Bartholdy C, et al. (2013) Qualitative and quantitative analysis of adenovirus type 5 vector-induced memory CD8 T cells: not as bad as their reputation. J Virol 87: 6283–6295.
    1. De Rosa SC, Thomas EP, Bui J, Huang Y, deCamp A, et al. (2011) HIV-DNA priming alters T cell responses to HIV-adenovirus vaccine even when responses to DNA are undetectable. J Immunol 187: 3391–3401.
    1. Kirman JR, Seder RA (2003) DNA vaccination: the answer to stable, protective T-cell memory? Curr Opin Immunol 15: 471–476.
    1. Schmidt NW, Podyminogin RL, Butler NS, Badovinac VP, Tucker BJ, et al. (2008) Memory CD8 T cell responses exceeding a large but definable threshold provide long-term immunity to malaria. Proc Natl Acad Sci U S A 105: 14017–14022.
    1. Quinn KM, Da Costa A, Yamamoto A, Berry D, Lindsay RW, et al. (2013) Comparative analysis of the magnitude, quality, phenotype, and protective capacity of simian immunodeficiency virus gag-specific CD8+ T cells following human-, simian-, and chimpanzee-derived recombinant adenoviral vector immunization. J Immunol 190: 2720–2735.
    1. Yang SH, Lee CG, Park SH, Im SJ, Kim YM, et al. (2006) Correlation of antiviral T-cell responses with suppression of viral rebound in chronic hepatitis B carriers: a proof-of-concept study. Gene Ther 13: 1110–1117.
    1. Penaloza-MacMaster P, Provine NM, Ra J, Borducchi EN, McNally A, et al. (2013) Alternative serotype adenovirus vaccine vectors elicit memory T cells with enhanced anamnestic capacity compared to Ad5 vectors. J Virol 87: 1373–1384.
    1. Tan WG, Jin HT, West EE, Penaloza-MacMaster P, Wieland A, et al. (2013) Comparative analysis of simian immunodeficiency virus gag-specific effector and memory CD8+ T cells induced by different adenovirus vectors. J Virol 87: 1359–1372.
    1. Todryk SM, Pathan AA, Keating S, Porter DW, Berthoud T, et al. (2009) The relationship between human effector and memory T cells measured by ex vivo and cultured ELISPOT following recent and distal priming. Immunology 128: 83–91.
    1. Huaman MC, Mullen GE, Long CA, Mahanty S (2009) Plasmodium falciparum apical membrane antigen 1 vaccine elicits multifunctional CD4 cytokine-producing and memory T cells. Vaccine 27: 5239–5246.
    1. Vuola JM, Keating S, Webster DP, Berthoud T, Dunachie S, et al. (2005) Differential immunogenicity of various heterologous prime-boost vaccine regimens using DNA and viral vectors in healthy volunteers. J Immunol 174: 449–455.
    1. Reyes-Sandoval A, Wyllie DH, Bauza K, Milicic A, Forbes EK, et al. (2011) CD8+ T Effector Memory Cells Protect against Liver-Stage Malaria. J Immunol 187: 1347–1357.
    1. Tse SW, Cockburn IA, Zhang H, Scott AL, Zavala F (2013) Unique transcriptional profile of liver-resident memory CD8+ T cells induced by immunization with malaria sporozoites. Genes Immun 14: 302–309.
    1. Cockburn IA, Chakravarty S, Overstreet MG, Garcia-Sastre A, Zavala F (2008) Memory CD8+ T cell responses expand when antigen presentation overcomes T cell self-regulation. J Immunol 180: 64–71.
    1. Hafalla JC, Morrot A, Sano G, Milon G, Lafaille JJ, et al. (2003) Early self-regulatory mechanisms control the magnitude of CD8+ T cell responses against liver stages of murine malaria. J Immunol 171: 964–970.
    1. Remarque EJ, Faber BW, Kocken CH, Thomas AW (2008) Apical membrane antigen 1: a malaria vaccine candidate in review. Trends Parasitol 24: 74–84.
    1. Sedegah M, Kim Y, Peters B, McGrath S, Ganeshan H, et al. (2011) Identification and localization of minimal MHC-restricted CD8+ T cell epitopes within the Plasmodium falciparum AMA1 protein. Malar J 9: 241.
    1. Elahi S, Dinges WL, Lejarcegui N, Laing KJ, Collier AC, et al. (2011) Protective HIV-specific CD8+ T cells evade Treg cell suppression. Nat Med 17: 989–995.

Source: PubMed

3
Předplatit