Vaccine Strain-Specificity of Protective HLA-Restricted Class 1 P. falciparum Epitopes

Martha Sedegah, Bjoern Peters, Michael R Hollingdale, Harini D Ganeshan, Jun Huang, Fouzia Farooq, Maria N Belmonte, Arnel D Belmonte, Keith J Limbach, Carter Diggs, Lorraine Soisson, Ilin Chuang, Eileen D Villasante, Martha Sedegah, Bjoern Peters, Michael R Hollingdale, Harini D Ganeshan, Jun Huang, Fouzia Farooq, Maria N Belmonte, Arnel D Belmonte, Keith J Limbach, Carter Diggs, Lorraine Soisson, Ilin Chuang, Eileen D Villasante

Abstract

A DNA prime/adenovirus boost malaria vaccine encoding Plasmodium falciparum strain 3D7 CSP and AMA1 elicited sterile clinical protection associated with CD8+ T cell interferon-gamma (IFN-γ) cells responses directed to HLA class 1-restricted AMA1 epitopes of the vaccine strain 3D7. Since a highly effective malaria vaccine must be broadly protective against multiple P. falciparum strains, we compared these AMA1 epitopes of two P. falciparum strains (7G8 and 3D7), which differ by single amino acid substitutions, in their ability to recall CD8+ T cell activities using ELISpot and flow cytometry/intracellular staining assays. The 7G8 variant peptides did not recall 3D7 vaccine-induced CD8+ T IFN-γ cell responses in these assays, suggesting that protection may be limited to the vaccine strain. The predicted MHC binding affinities of the 7G8 variant epitopes were similar to the 3D7 epitopes, suggesting that the amino acid substitutions of the 7G8 variants may have interfered with TCR recognition of the MHC:peptide complex or that the 7G8 variant may have acted as an altered peptide ligand. These results stress the importance of functional assays in defining protective epitopes. Clinical Trials Registrations: NCT00870987, NCT00392015.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. ELISpot and CD8+ T cell…
Fig 1. ELISpot and CD8+ T cell IFN-γ responses of DNA/HuAd5 and HuAd5 immunized subjects to P. falciparum strains 3D7 and 7G8 AMA1 A*03 protective epitopes.
ELISpot and CD8+ T cell IFN-γ activities are shown in Panels A–D. Panel A: ELISpot IFN-γ response of the A*03 protected subject (v11) are positive with Ap8 and the 3D7 A*03 epitope but not the 7G8 epitope (arrow). Panel B: ELISpot activity of v11 is not affected by CD4+-depletion but is abolished after CD8+ depletion (arrow). Panel C: CD8+ T cell IFN-γ responses of v11 are much higher (p = 0.001) to the 3D7 epitope than to the 7G8 epitope (arrow). Panel D: ELISpot IFN-γ responses of two of four non-protected subjects from the HuAd5 trial were weakly positive with the 3D7 epitope but all four subjects were negative with the 7G8 epitope (arrows).
Fig 2. ELISpot and CD8+ T cell…
Fig 2. ELISpot and CD8+ T cell IFN-γ responses of DNA/HuAd5 and HuAd5 immunized subjects to P. falciparum strains 3D7 and 7G8 AMA1 B*58 protective epitopes.
ELISpot and CD8+ T cell IFN-γ activities are shown in Panels A–D. Panel A: ELISpot IFN-γ responses of the B*58 protected subjects (v10, v18) are positive with Ap10 and the 3D7 B*58 epitope but not 7G8 epitopes (arrows). Panel B: ELISpot activity of v10 is reduced but still remains positive after CD4+-depletion, but is abolished after CD8+-depletion (arrow); activity of v18 is unaffected by CD4+-depletion but is abolished after CD8+-depletion (arrow). Panel C: CD8+ T cell responses of v10 and v18 are much higher (p = 0.001) against the 3D7 B*58 epitope rather than the 7G8 B*58 epitope (arrows). Panel D: ELISpot IFN-γ response of DNA/HuAd5 non-protected B*58 subject (v15) was negative and non-protected v194 was weakly positive with the 3D7 B*58 epitope; two non-protected subjects from the HuAd5 trial that express A*01 (v135, v179) were weakly positive with the 3D7 B*58 epitope; all these subjects were negative with the 7G8 B*58 epitope (arrows).

References

    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 10.1371/journal.pone.0055571
    1. Tamminga C, Sedegah M, Maiolatesi S, Fedders C, Reyes S, Reyes A, et al. 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. 2013;9:2165–77. 10.4161/hv.24941
    1. Sedegah M, Hollingdale MR, Farooq F, Ganeshan H, Belmonte M, Kim Y, et al. Sterile Immunity to Malaria after DNA Prime/Adenovirus Boost Immunization Is Associated with Effector Memory CD8+T Cells Targeting AMA1 Class I Epitopes. PLoS One. 2014;9:e106241 10.1371/journal.pone.0106241
    1. Lundegaard C, Lamberth K, Harndahl M, Buus S, Lund O, Nielsen M. NetMHC-3.0: accurate web accessible predictions of human, mouse and monkey MHC class I affinities for peptides of length 8–11. Nucleic Acids Res. 2008;36:W509–12. 10.1093/nar/gkn202
    1. Barry AE, Arnott A. Strategies for designing and monitoring malaria vaccines targeting diverse antigens. Front Immunol. 2014;5:359 10.3389/fimmu.2014.00359
    1. Remarque EJ, Faber BW, Kocken CH, Thomas AW. Apical membrane antigen 1: a malaria vaccine candidate in review. Trends Parasitol. 2008;24:74–84. 10.1016/j.pt.2007.12.002
    1. Drew DR, Hodder AN, Wilson DW, Foley M, Mueller I, Siba PM, et al. Defining the antigenic diversity of Plasmodium falciparum apical membrane antigen 1 and the requirements for a multi-allele vaccine against malaria. PLoS One. 2012;7:e51023 10.1371/journal.pone.0051023
    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 10.1371/journal.pone.0024586
    1. Nielsen M, Lundegaard C, Blicher T, Lamberth K, Harndahl M, Justesen S, et al. NetMHCpan, a method for quantitative predictions of peptide binding to any HLA-A and -B locus protein of known sequence. PLoS One. 2007;2:e796 10.1371/journal.pone.0000796
    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. 10.1073/pnas.181123498
    1. Stewart VA, McGrath SM, Dubois PM, Pau MG, Mettens P, Shott J, 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;75:2283–90. 10.1128/IAI.01879-06
    1. Lundegaard C, Lund O, Nielsen M. 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. 2008;24:1397–8. 10.1093/bioinformatics/btn128
    1. Sidney J, Peters B, Frahm N, Brander C, Sette A. HLA class I supertypes: a revised and updated classification. BMC Immunol. 2008;9:1 10.1186/1471-2172-9-1
    1. Garcia KC, Degano M, Stanfield RL, Brunmark A, Jackson MR, Peterson PA, et al. An alphabeta T cell receptor structure at 2.5 A and its orientation in the TCR-MHC complex. Science. 1996;274:209–19. 10.1126/science.274.5285.209
    1. Hennecke J, Carfi A, Wiley DC. Structure of a covalently stabilized complex of a human alphabeta T-cell receptor, influenza HA peptide and MHC class II molecule, HLA-DR1. EMBO J. 2000;19:5611–24. 10.1093/emboj/19.21.5611
    1. Marrack P, Scott-Browne JP, Dai S, Gapin L, Kappler JW. Evolutionarily conserved amino acids that control TCR-MHC interaction. Annu Rev Immunol. 2008;26:171–203. 10.1146/annurev.immunol.26.021607.090421
    1. Hawse WF, De S, Greenwood AI, Nicholson LK, Zajicek J, Kovrigin EL, et al. TCR scanning of peptide/MHC through complementary matching of receptor and ligand molecular flexibility. J Immunol. 2014;192:2885–91. 10.4049/jimmunol.1302953
    1. Pircher H, Moskophidis D, Rohrer U, Burki K, Hengartner H, Zinkernagel RM. Viral escape by selection of cytotoxic T cell-resistant virus variants in vivo. Nature. 1990;346:629–33. 10.1038/346629a0
    1. Bermudez A, Calderon D, Moreno-Vranich A, Almonacid H, Patarroyo MA, Poloche A, et al. Gauche(+) side-chain orientation as a key factor in the search for an immunogenic peptide mixture leading to a complete fully protective vaccine. Vaccine. 2014;32:2117–26. 10.1016/j.vaccine.2014.02.003
    1. Patarroyo ME, Bermudez A, Alba MP, Vanegas M, Moreno-Vranich A, Poloche LA, et al. IMPIPS: the immune protection-inducing protein structure concept in the search for steric-electron and topochemical principles for complete fully-protective chemically synthesised vaccine development. PLoS One. 2015;10:e0123249 10.1371/journal.pone.0123249
    1. Valkenburg SA, Gras S, Guillonneau C, Hatton LA, Bird NA, Twist KA, et al. Preemptive priming readily overcomes structure-based mechanisms of virus escape. Proc Natl Acad Sci U S A. 2013;110:5570–5. 10.1073/pnas.1302935110
    1. Valkenburg SA, Quinones-Parra S, Gras S, Komadina N, McVernon J, Wang Z, et al. Acute emergence and reversion of influenza A virus quasispecies within CD8+ T cell antigenic peptides. Nat Commun. 2013;4:2663 10.1038/ncomms3663
    1. Bronke C, Almeida CA, McKinnon E, Roberts SG, Keane NM, Chopra A, et al. HIV escape mutations occur preferentially at HLA-binding sites of CD8 T-cell epitopes. AIDS. 2013;27:899–905. 10.1097/QAD.0b013e32835e1616
    1. Plebanski M, Lee EA, Hill AV. Immune evasion in malaria: altered peptide ligands of the circumsporozoite protein. Parasitology. 1997;115 Suppl:S55–66. 10.1017/s0031182097002035
    1. Flanagan KL, Wilson KL, Plebanski M. Polymorphism in liver-stage malaria vaccine candidate proteins: immune evasion and implications for vaccine design. Expert Rev Vaccines. 2016;15:389–99. 10.1586/14760584.2016.1125785
    1. Wilson KL, Xiang SD, Plebanski M. A Model to Study the Impact of Polymorphism Driven Liver-Stage Immune Evasion by Malaria Parasites, to Help Design Effective Cross-Reactive Vaccines. Front Microbiol. 2016;7:303 10.3389/fmicb.2016.00303
    1. Gilbert SC, Plebanski M, Gupta S, Morris J, Cox M, Aidoo M, et al. Association of malaria parasite population structure, HLA, and immunological antagonism. Science. 1998;279:1173–7. 10.1126/science.279.5354.1173
    1. Mantzourani ED, Platts JA, Brancale A, Mavromoustakos TM, Tselios TV. Molecular dynamics at the receptor level of immunodominant myelin basic protein epitope 87–99 implicated in multiple sclerosis and its antagonists altered peptide ligands: triggering of immune response. J Mol Graph Model. 2007;26:471–81. 10.1016/j.jmgm.2007.02.004
    1. Fadda L, Borhis G, Ahmed P, Cheent K, Pageon SV, Cazaly A, et al. Peptide antagonism as a mechanism for NK cell activation. Proc Natl Acad Sci U S A. 2010;107:10160–5. 10.1073/pnas.0913745107
    1. Kirkman LA, Deitsch KW. Recombination and Diversification of the Variant Antigen Encoding Genes in the Malaria Parasite Plasmodium falciparum. Microbiol Spectr. 2014;2 10.1128/microbiolspec.MDNA3-0022-2014
    1. Terheggen U, Drew DR, Hodder AN, Cross NJ, Mugyenyi CK, Barry AE, et al. Limited antigenic diversity of Plasmodium falciparum apical membrane antigen 1 supports the development of effective multi-allele vaccines. BMC Med. 2014;12:183 10.1186/s12916-014-0183-5
    1. Thera MA, Doumbo OK, Coulibaly D, Laurens MB, Ouattara A, Kone AK, et al. A field trial to assess a blood-stage malaria vaccine. N Engl J Med. 2011;365:1004–13. 10.1056/NEJMoa1008115
    1. Coley AM, Parisi K, Masciantonio R, Hoeck J, Casey JL, Murphy VJ, et al. The most polymorphic residue on Plasmodium falciparum apical membrane antigen 1 determines binding of an invasion-inhibitory antibody. Infect Immun. 2006;74:2628–36. 10.1128/IAI.74.5.2628-2636.2006
    1. Elias SC, Collins KA, Halstead FD, Choudhary P, Bliss CM, Ewer KJ, et al. Assessment of immune interference, antagonism, and diversion following human immunization with biallelic blood-stage malaria viral-vectored vaccines and controlled malaria infection. J Immunol. 2013;190:1135–47. 10.4049/jimmunol.1201455
    1. Lee EA, Flanagan KL, Minigo G, Reece WH, Bailey R, Pinder M, et al. Dimorphic Plasmodium falciparum merozoite surface protein-1 epitopes turn off memory T cells and interfere with T cell priming. Eur J Immunol. 2006;36:1168–78. 10.1002/eji.200526010
    1. Plebanski M, Lee EA, Hannan CM, Flanagan KL, Gilbert SC, Gravenor MB, et al. Altered peptide ligands narrow the repertoire of cellular immune responses by interfering with T-cell priming. Nat Med. 1999;5:565–71. 10.1038/8444
    1. Ewer KJ, O'Hara GA, Duncan CJ, Collins KA, Sheehy SH, Reyes-Sandoval A, et al. Protective CD8+ T-cell immunity to human malaria induced by chimpanzee adenovirus-MVA immunisation. Nat Commun. 2013;4:2836 10.1038/ncomms3836
    1. Hodgson SH, Juma E, Salim A, Magiri C, Kimani D, Njenga D, et al. Evaluating controlled human malaria infection in Kenyan adults with varying degrees of prior exposure to Plasmodium falciparum using sporozoites administered by intramuscular injection. Front Microbiol. 2014;5:686 10.3389/fmicb.2014.00686
    1. Sedegah M, Kim Y, Peters B, McGrath S, Ganeshan H, Lejano J, et al. Identification and localization of minimal MHC-restricted CD8+ T cell epitopes within the Plasmodium falciparum AMA1 protein. Malar J. 2011;9:241 10.1186/1475-2875-9-241
    1. Sedegah M, Kim D, Ganeshan H, Huang J, Belmonte M, Abot E, et al. Identification of minimal human MHC-restricted CD8+ T-cell epitopes within the Plasmodium falciparum circumsporozoite protein (CSP). Malar J. 2013. 10.1186/1475-2875-12-185
    1. Doolan DL, Southwood S, Chesnut R, Appella E, Gomez E, Richards A, 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;165:1123–37. 10.4049/jimmunol.165.2.1123
    1. Sette A, Sidney J. Nine major HLA class I supertypes account for the vast preponderance of HLA-A and -B polymorphism. Immunogenetics. 1999;50:201–12. 10.1007/s002510050594
    1. Seder RA, Chang LJ, Enama ME, Zephir KL, Sarwar UN, Gordon IJ, et al. Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science. 2013;341:1359–65. 10.1126/science.1241800

Source: PubMed

3
Tilaa