Identification and Immune Assessment of T Cell Epitopes in Five Plasmodium falciparum Blood Stage Antigens to Facilitate Vaccine Candidate Selection and Optimization

The hurdles to effective blood stage malaria vaccine design include immune evasion tactics used by the parasite such as redundant invasion pathways and antigen variation among circulating parasite strains. While blood stage malaria vaccine development primarily focuses on eliciting optimal humoral responses capable of blocking erythrocyte invasion, clinically-tested Plasmodium falciparum (Pf) vaccines have not elicited sterile protection, in part due to the dramatically high levels of antibody needed. Recent development efforts with non-redundant, conserved blood stage antigens suggest both high antibody titer and rapid antibody binding kinetics are important efficacy factors. Based on the central role of helper CD4 T cells in development of strong, protective immune responses, we systematically analyzed the class II epitope content in five leading Pf blood stage antigens (RH5, CyRPA, RIPR, AMA1 and EBA175) using in silico, in vitro, and ex vivo methodologies. We employed in silico T cell epitope analysis to enable identification of 67 HLA-restricted class II epitope clusters predicted to bind a panel of nine HLA-DRB1 alleles. We assessed a subset of these for HLA-DRB1 allele binding in vitro, to verify the in silico predictions. All clusters assessed (40 clusters represented by 46 peptides) bound at least two HLA-DR alleles in vitro. The overall epitope prediction to in vitro HLA-DRB1 allele binding accuracy was 71%. Utilizing the set of RH5 class II epitope clusters (10 clusters represented by 12 peptides), we assessed stimulation of T cells collected from HLA-matched RH5 vaccinees using an IFN-γ T cell recall assay. All clusters demonstrated positive recall responses, with the highest responses – by percentage of responders and response magnitude – associated with clusters located in the N-terminal region of RH5. Finally, a statistically significant correlation between in silico epitope predictions and ex vivo IFN-γ recall response was found when accounting for HLA-DR matches between the epitope predictions and donor HLA phenotypes. This is the first comprehensive analysis of class II epitope content in RH5, CyRPA, RIPR, AMA1 and EBA175 accompanied by in vitro HLA binding validation for all five proteins and ex vivo T cell response confirmation for RH5.

[1]  G. Alter,et al.  Reduced blood-stage malaria growth and immune correlates in humans following RH5 vaccination , 2021, Med.

[2]  B. Haynes,et al.  Protein/AS01B vaccination elicits stronger, more Th2-skewed antigen-specific human T follicular helper cell responses than heterologous viral vectors , 2021, Cell reports. Medicine.

[3]  A. Dent,et al.  T Helper Plasticity Is Orchestrated by STAT3, Bcl6, and Blimp-1 Balancing Pathology and Protection in Malaria , 2020, iScience.

[4]  M. Higgins,et al.  The RH5-CyRPA-Ripr Complex as a Malaria Vaccine Target , 2020, Trends in parasitology.

[5]  Guilhem Richard,et al.  Better Epitope Discovery, Precision Immune Engineering, and Accelerated Vaccine Design Using Immunoinformatics Tools , 2020, Frontiers in Immunology.

[6]  S. Kappe,et al.  Human Antibodies that Slow Erythrocyte Invasion Potentiate Malaria-Neutralizing Antibodies , 2019, Cell.

[7]  Z. Zenonos,et al.  A defined mechanistic correlate of protection against Plasmodium falciparum malaria in non-human primates , 2019, Nature Communications.

[8]  A. Cowman,et al.  Neutralising antibodies block the function of Rh5/Ripr/CyRPA complex during invasion of Plasmodium falciparum into human erythrocytes , 2019, Cellular microbiology.

[9]  A. D. De Groot,et al.  Immune escape and immune camouflage may reduce the efficacy of RTS,S vaccine in Malawi , 2019, Human vaccines & immunotherapeutics.

[10]  K. Kedzierska,et al.  With a Little Help from T Follicular Helper Friends: Humoral Immunity to Influenza Vaccination , 2019, The Journal of Immunology.

[11]  Alessandro Sette,et al.  The Immune Epitope Database (IEDB): 2018 update , 2018, Nucleic Acids Res..

[12]  A. Lawrie,et al.  Production, quality control, stability, and potency of cGMP-produced Plasmodium falciparum RH5.1 protein vaccine expressed in Drosophila S2 cells , 2018, npj Vaccines.

[13]  J. Lieberman,et al.  Cytotoxic CD8+ T cells recognize and kill Plasmodium vivax-infected reticulocytes , 2018, Nature Medicine.

[14]  Peter D. Crompton,et al.  Atypical activation of dendritic cells by Plasmodium falciparum , 2017, Proceedings of the National Academy of Sciences.

[15]  Juliana K. Wambua,et al.  Human vaccination against RH5 induces neutralizing antimalarial antibodies that inhibit RH5 invasion complex interactions. , 2017, JCI insight.

[16]  E. Tartour,et al.  Safety and immunogenicity of a recombinant Plasmodium falciparum AMA1-DiCo malaria vaccine adjuvanted with GLA-SE or Alhydrogel® in European and African adults: A phase 1a/1b, randomized, double-blind multi-centre trial. , 2017, Vaccine.

[17]  J. McCarthy,et al.  Human vaccination against Plasmodium vivax Duffy-binding protein induces strain-transcending antibodies , 2017, JCI insight.

[18]  Arnone Nithichanon,et al.  A humanized mouse model identifies key amino acids for low immunogenicity of H7N9 vaccines , 2017, Scientific Reports.

[19]  J. Ocran,et al.  Safety and Immunogenicity of EBA-175 RII-NG Malaria Vaccine Administered Intramuscularly in Semi-Immune Adults: A Phase 1, Double-Blinded Placebo Controlled Dosage Escalation Study , 2016, PloS one.

[20]  P. Borrow,et al.  Germinal Center B Cell and T Follicular Helper Cell Responses to Viral Vector and Protein-in-Adjuvant Vaccines , 2016, The Journal of Immunology.

[21]  L. Ochola-Oyier,et al.  Implications from predicted B-cell and T-cell epitopes of Plasmodium falciparum merozoite proteins EBA175-RII and Rh5 , 2016, Bioinformation.

[22]  C. Poh,et al.  Mice lacking Programmed cell death-1 show a role for CD8+ T cells in long-term immunity against blood-stage malaria , 2016, Scientific Reports.

[23]  Qingsheng Li,et al.  Vaccine Induction of Lymph Node–Resident Simian Immunodeficiency Virus Env-Specific T Follicular Helper Cells in Rhesus Macaques , 2016, The Journal of Immunology.

[24]  C. Leslie,et al.  A mechanism for expansion of regulatory T cell repertoire and its role in self tolerance , 2015, Nature.

[25]  Leonard Moise,et al.  iVAX: An integrated toolkit for the selection and optimization of antigens and the design of epitope-driven vaccines , 2015, Human vaccines & immunotherapeutics.

[26]  A. D. De Groot,et al.  H7N9 T-cell epitopes that mimic human sequences are less immunogenic and may induce Treg-mediated tolerance , 2015, Human vaccines & immunotherapeutics.

[27]  J. Rayner,et al.  Plasmodium falciparum Erythrocyte Invasion: Combining Function with Immune Evasion , 2014, PLoS pathogens.

[28]  Michel Theron,et al.  Neutralization of Plasmodium falciparum Merozoites by Antibodies against PfRH5 , 2014, The Journal of Immunology.

[29]  R. Ahmed,et al.  PD-1 dependent exhaustion of CD8+ T cells drives chronic malaria. , 2013, Cell reports.

[30]  A. D. De Groot,et al.  Universal H1N1 influenza vaccine development , 2013, Human vaccines & immunotherapeutics.

[31]  A. Rothman,et al.  The two-faced T cell epitope , 2013, Human vaccines & immunotherapeutics.

[32]  Virginia Pascual,et al.  Induction of ICOS+CXCR3+CXCR5+ TH Cells Correlates with Antibody Responses to Influenza Vaccination , 2013, Science Translational Medicine.

[33]  R. Cortese,et al.  ChAd63-MVA-vectored blood-stage malaria vaccines targeting MSP1 and AMA1: assessment of efficacy against mosquito bite challenge in humans. , 2012, Molecular therapy : the journal of the American Society of Gene Therapy.

[34]  R. Cortese,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 , 2012, PloS one.

[35]  R. Cortese,et al.  Phase Ia clinical evaluation of the Plasmodium falciparum blood-stage antigen MSP1 in ChAd63 and MVA vaccine vectors. , 2011, Molecular therapy : the journal of the American Society of Gene Therapy.

[36]  O. Doumbo,et al.  A field trial to assess a blood-stage malaria vaccine. , 2011, The New England journal of medicine.

[37]  Anne S De Groot,et al.  Coupling sensitive in vitro and in silico techniques to assess cross-reactive CD4(+) T cells against the swine-origin H1N1 influenza virus. , 2011, Vaccine.

[38]  W. Keitel,et al.  Safety and Immunogenicity of a Recombinant Nonglycosylated Erythrocyte Binding Antigen 175 Region II Malaria Vaccine in Healthy Adults Living in an Area Where Malaria Is Not Endemic , 2010, Clinical and Vaccine Immunology.

[39]  S. Draper,et al.  Blood-stage malaria vaccines — recent progress and future challenges , 2010, Annals of tropical medicine and parasitology.

[40]  J. Barnwell,et al.  High Antibody Titer against Apical Membrane Antigen-1 Is Required to Protect against Malaria in the Aotus Model , 2009, PloS one.

[41]  A. D. De Groot,et al.  T cell epitope: Friend or Foe? Immunogenicity of biologics in context☆ , 2009, Advanced Drug Delivery Reviews.

[42]  C. Engwerda,et al.  Recent insights into humoral and cellular immune responses against malaria. , 2008, Trends in parasitology.

[43]  William W. Kwok,et al.  Antibiotic-refractory Lyme arthritis is associated with HLA-DR molecules that bind a Borrelia burgdorferi peptide , 2006, The Journal of experimental medicine.

[44]  M. Kenzelmann,et al.  Argonaute—a database for gene regulation by mammalian microRNAs , 2005, BMC Bioinformatics.

[45]  D. Kioussis,et al.  Malaria-specific transgenic CD4(+) T cells protect immunodeficient mice from lethal infection and demonstrate requirement for a protective threshold of antibody production for parasite clearance. , 2005, Blood.

[46]  Ellis L. Reinherz,et al.  PEPVAC: a web server for multi-epitope vaccine development based on the prediction of supertypic MHC ligands , 2005, Nucleic Acids Res..

[47]  Markus S. Mueller,et al.  A Role for Apical Membrane Antigen 1 during Invasion of Hepatocytes by Plasmodium falciparum Sporozoites* , 2004, Journal of Biological Chemistry.

[48]  O. Lund,et al.  Definition of supertypes for HLA molecules using clustering of specificity matrices , 2004, Immunogenetics.

[49]  A. Ménez,et al.  Complementarity and redundancy of the binding specificity of HLA‐DRB1, ‐DRB3, ‐DRB4 and ‐DRB5 molecules , 2001, European journal of immunology.

[50]  J. Sidney,et al.  Nine major HLA class I supertypes account for the vast preponderance of HLA-A and -B polymorphism , 1999, Immunogenetics.

[51]  M F del Guercio,et al.  Several common HLA-DR types share largely overlapping peptide binding repertoires. , 1998, Journal of immunology.

[52]  C. Bailey-Kellogg,et al.  HCV epitope, homologous to multiple human protein sequences, induces a regulatory T cell response in infected patients. , 2015, Journal of hepatology.