1
|
Thiam LG, McHugh K, Ba A, Li R, Guo Y, Pouye MN, Cisse A, Pipini D, Diallo F, Sene SD, Patel SD, Thiam A, Sadio BD, Mbengue A, Vigan-Womas I, Sheng Z, Shapiro L, Draper SJ, Bei AK. Vaccine-induced human monoclonal antibodies to PfRH5 show broadly neutralizing activity against P. falciparum clinical isolates. NPJ Vaccines 2024; 9:198. [PMID: 39448626 PMCID: PMC11502735 DOI: 10.1038/s41541-024-00986-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/28/2024] [Indexed: 10/26/2024] Open
Abstract
Vaccines to the Plasmodium falciparum reticulocyte binding-like protein homologue 5 (PfRH5) target the blood-stage of the parasite life cycle. PfRH5 has the potential to trigger the production of strain-transcendent antibodies and has proven its efficacy both in pre-clinical and early clinical studies. Vaccine-induced monoclonal antibodies (mAbs) to PfRH5 showed promising outcomes against cultured P. falciparum laboratory strains from distinct geographic areas. Here, we assessed the functional impact of vaccine-induced anti-PfRH5 mAbs on more genetically diverse P. falciparum clinical isolates. We used mAbs previously isolated from single B cells of UK adult PfRH5 vaccinees and used ex-vivo growth inhibition activity (GIA) assays to assess their efficacy against P. falciparum clinical isolates. Next-generation sequencing (NGS) was used to assess the breadth of genetic diversity in P. falciparum clinical isolates and to infer the genotype/phenotype relationship involved in antibody susceptibility. We showed a dose-dependent inhibition of clinical isolates with three main GIA groups: high, medium and low. Except for one isolate, our data show no significant differences in the mAb GIA profile between P. falciparum clinical isolates and the 3D7 reference strain, which harbors the vaccine allele. We also observed an additive relationship for mAb combinations, whereby the combination of GIA-low and GIA-medium antibodies resulted in increased GIA, having important implications for the contribution of specific clones within polyclonal IgG responses. While our NGS analysis showed the occurrence of novel mutations in the pfrh5 gene, these mutations were predicted to have little or no functional impact on the antigen structure or recognition by known mAbs. Our present findings complement earlier reports on the strain transcendent potential of anti-PfRH5 mAbs and constitute, to our knowledge, the first report on the susceptibility of P. falciparum clinical isolates from natural infections to vaccine-induced human mAbs to PfRH5.
Collapse
Affiliation(s)
- Laty G Thiam
- G4 - Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Kirsty McHugh
- Department of Biochemistry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Aboubacar Ba
- G4 - Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Rebecca Li
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Yicheng Guo
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Mariama N Pouye
- G4 - Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Awa Cisse
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Dimitra Pipini
- Department of Biochemistry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Fatoumata Diallo
- G4 - Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Seynabou D Sene
- G4 - Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Saurabh D Patel
- G4 - Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Alassane Thiam
- G4 - Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Bacary D Sadio
- Pôle Virologie, Institut Pasteur de Dakar, Dakar, Senegal
| | - Alassane Mbengue
- G4 - Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Inés Vigan-Womas
- G4 - Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Zizhang Sheng
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Lawrence Shapiro
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Biochemistry and Biophysics, Columbia University, New York, NY, USA
| | - Simon J Draper
- Department of Biochemistry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Amy K Bei
- G4 - Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal.
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA.
| |
Collapse
|
2
|
Duffy PE, Gorres JP, Healy SA, Fried M. Malaria vaccines: a new era of prevention and control. Nat Rev Microbiol 2024:10.1038/s41579-024-01065-7. [PMID: 39025972 DOI: 10.1038/s41579-024-01065-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2024] [Indexed: 07/20/2024]
Abstract
Malaria killed over 600,000 people in 2022, a death toll that has not improved since 2015. Additionally, parasites and mosquitoes resistant to existing interventions are spreading across Africa and other regions. Vaccines offer hope to reduce the mortality burden: the first licensed malaria vaccines, RTS,S and R21, will be widely deployed in 2024 and should substantially reduce childhood deaths. In this Review, we provide an overview of the malaria problem and the Plasmodium parasite, then describe the RTS,S and R21 vaccines (the first vaccines for any human parasitic disease), summarizing their benefits and limitations. We explore next-generation vaccines designed using new knowledge of malaria pathogenesis and protective immunity, which incorporate antigens and platforms to elicit effective immune responses against different parasite stages in human or mosquito hosts. We describe a decision-making process that prioritizes malaria vaccine candidates for development in a resource-constrained environment. Future vaccines might improve upon the protective efficacy of RTS,S or R21 for children, or address the wider malaria scourge by preventing pregnancy malaria, reducing the burden of Plasmodium vivax or accelerating malaria elimination.
Collapse
Affiliation(s)
- Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - J Patrick Gorres
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sara A Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michal Fried
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
3
|
Miura K, Flores-Garcia Y, Long CA, Zavala F. Vaccines and monoclonal antibodies: new tools for malaria control. Clin Microbiol Rev 2024; 37:e0007123. [PMID: 38656211 PMCID: PMC11237600 DOI: 10.1128/cmr.00071-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024] Open
Abstract
SUMMARYMalaria remains one of the biggest health problems in the world. While significant reductions in malaria morbidity and mortality had been achieved from 2000 to 2015, the favorable trend has stalled, rather significant increases in malaria cases are seen in multiple areas. In 2022, there were 249 million estimated cases, and 608,000 malaria-related deaths, mostly in infants and children aged under 5 years, globally. Therefore, in addition to the expansion of existing anti-malarial control measures, it is critical to develop new tools, such as vaccines and monoclonal antibodies (mAbs), to fight malaria. In the last 2 years, the first and second malaria vaccines, both targeting Plasmodium falciparum circumsporozoite proteins (PfCSP), have been recommended by the World Health Organization to prevent P. falciparum malaria in children living in moderate to high transmission areas. While the approval of the two malaria vaccines is a considerable milestone in vaccine development, they have much room for improvement in efficacy and durability. In addition to the two approved vaccines, recent clinical trials with mAbs against PfCSP, blood-stage vaccines against P. falciparum or P. vivax, and transmission-blocking vaccine or mAb against P. falciparum have shown promising results. This review summarizes the development of the anti-PfCSP vaccines and mAbs, and recent topics in the blood- and transmission-blocking-stage vaccine candidates and mAbs. We further discuss issues of the current vaccines and the directions for the development of next-generation vaccines.
Collapse
Affiliation(s)
- Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Malaria Research Institute, Baltimore, Maryland, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Malaria Research Institute, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Nkumama IN, Ogwang R, Odera D, Musasia F, Mwai K, Nyamako L, Murungi L, Tuju J, Fürle K, Rosenkranz M, Kimathi R, Njuguna P, Hamaluba M, Kapulu MC, Frank R, Osier FHA. Breadth of Fc-mediated effector function correlates with clinical immunity following human malaria challenge. Immunity 2024; 57:1215-1224.e6. [PMID: 38788711 PMCID: PMC7616646 DOI: 10.1016/j.immuni.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/19/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024]
Abstract
Malaria is a life-threatening disease of global health importance, particularly in sub-Saharan Africa. The growth inhibition assay (GIA) is routinely used to evaluate, prioritize, and quantify the efficacy of malaria blood-stage vaccine candidates but does not reliably predict either naturally acquired or vaccine-induced protection. Controlled human malaria challenge studies in semi-immune volunteers provide an unparalleled opportunity to robustly identify mechanistic correlates of protection. We leveraged this platform to undertake a head-to-head comparison of seven functional antibody assays that are relevant to immunity against the erythrocytic merozoite stage of Plasmodium falciparum. Fc-mediated effector functions were strongly associated with protection from clinical symptoms of malaria and exponential parasite multiplication, while the gold standard GIA was not. The breadth of Fc-mediated effector function discriminated clinical immunity following the challenge. These findings present a shift in the understanding of the mechanisms that underpin immunity to malaria and have important implications for vaccine development.
Collapse
Affiliation(s)
- Irene N Nkumama
- Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany; Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya; European Vaccine Initiative, Heidelberg, Germany
| | - Rodney Ogwang
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya
| | - Dennis Odera
- Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany; Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya
| | - Fauzia Musasia
- Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Kennedy Mwai
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya; Epidemiology and Biostatistics Division, School of Public Health, University of the Witwatersrand, Johannesburg, South Africa
| | - Lydia Nyamako
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya
| | - Linda Murungi
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya
| | - James Tuju
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya; Department of Biotechnology and Biochemistry, Pwani University, Kilifi, Kenya
| | - Kristin Fürle
- Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Micha Rosenkranz
- Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Rinter Kimathi
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya
| | - Patricia Njuguna
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya
| | - Mainga Hamaluba
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya
| | - Melissa C Kapulu
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya
| | - Roland Frank
- Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Faith H A Osier
- Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany; Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya; Department of Life Sciences, Imperial College London, London, UK.
| |
Collapse
|
5
|
Miura K. How to Accelerate Early Stage of Malaria Vaccine Development by Optimizing Functional Assays. Vaccines (Basel) 2024; 12:586. [PMID: 38932315 PMCID: PMC11209467 DOI: 10.3390/vaccines12060586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
While two Plasmodium falciparum circumsporozoite protein-based pre-erythrocytic vaccines (PEV), RTS,S and R21, have been approved by the WHO, no blood-stage vaccine (BSV) or transmission-blocking vaccine (TBV) has reached a phase 3 trial. One of the major obstacles that slows down malaria vaccine development is the shortage (or lack) of in vitro assays or animal models by which investigators can reasonably select the best vaccine formulation (e.g., antigen, adjuvant, or platform) and/or immunization strategy (e.g., interval of inoculation or route of immunization) before a human phase 2 trial. In the case of PEV, RTS,S and R21 have set a benchmark, and a new vaccine can be compared with (one of) the approved PEV directly in preclinical or early clinical studies. However, such an approach cannot be utilized for BSV or TBV development at this moment. The focus of this review is in vitro assays or in vivo models that can be used for P. falciparum BSV or TBV development, and I discuss important considerations during assay selection, standardization, qualification, validation, and interpretation of the assay results. Establishment of a robust assay/model with proper interpretation of the results is the one of key elements to accelerate future vaccine development.
Collapse
Affiliation(s)
- Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
6
|
Cummings JF, Polhemus ME, Kester KE, Ockenhouse CF, Gasser RA, Coyne P, Wortmann G, Nielsen RK, Schaecher K, Holland CA, Krzych U, Tornieporth N, Soisson LA, Angov E, Heppner DG. A phase IIa, randomized, double-blind, safety, immunogenicity and efficacy trial of Plasmodium falciparum vaccine antigens merozoite surface protein 1 and RTS,S formulated with AS02 adjuvant in healthy, malaria-naïve adults. Vaccine 2024; 42:3066-3074. [PMID: 38584058 DOI: 10.1016/j.vaccine.2024.03.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 03/15/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND To improve the efficacy of Plasmodium falciparum malaria vaccine RTS,S/AS02, we conducted a study in 2001 in healthy, malaria-naïve adults administered RTS,S/AS02 in combination with FMP1, a recombinant merozoite surface-protein-1, C-terminal 42kD fragment. METHODS A double-blind Phase I/IIa study randomized N = 60 subjects 1:1:1:1 to one of four groups, N = 15/group, to evaluate safety, immunogenicity, and efficacy of intra-deltoid half-doses of RTS,S/AS02 and FMP1/AS02 administered in the contralateral (RTS,S + FMP1-separate) or same (RTS,S + FMP1-same) sites, or FMP1/AS02 alone (FMP1-alone), or RTS,S/AS02 alone (RTS,S-alone) on a 0-, 1-, 3-month schedule. Subjects receiving three doses of vaccine and non-immunized controls (N = 11) were infected with homologous P. falciparum 3D7 sporozoites by Controlled Human Malaria Infection (CHMI). RESULTS Subjects in all vaccination groups experienced mostly mild or moderate local and general adverse events that resolved within eight days. Anti-circumsporozoite antibody levels were lower when FMP1 and RTS,S were co-administered at the same site (35.0 µg/mL: 95 % CI 20.3-63), versus separate arms (57.4 µg/mL: 95 % CI 32.3-102) or RTS,S alone (62.0 µg/mL: 95 % CI: 37.8-101.8). RTS,S-specific lymphoproliferative responses and ex vivo ELISpot CSP-specific interferon-gamma (IFN-γ) responses were indistinguishable among groups receiving RTS,S/AS02. There was no difference in antibody to FMP1 among groups receiving FMP1/AS02. After CHMI, groups immunized with a RTS,S-containing regimen had ∼ 30 % sterile protection against parasitemia, and equivalent delays in time-to-parasitemia. The FMP1/AS02 alone group showed no sterile immunity or delay in parasitemia. CONCLUSION Co-administration of RTS,S and FMP1/AS02 reduced anti-RTS,S antibody, but did not affect tolerability, cellular immunity, or efficacy in a stringent CHMI model. Absence of efficacy or delay of patency in the sporozoite challenge model in the FMP1/AS02 group did not rule out efficacy of FMP1/AS02 in an endemic population. However, a Phase IIb trial of FMP1/AS02 in children in malaria-endemic Kenya did not demonstrate efficacy against natural infection. CLINICALTRIALS gov identifier: NCT01556945.
Collapse
Affiliation(s)
- J F Cummings
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - M E Polhemus
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - K E Kester
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - C F Ockenhouse
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - R A Gasser
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - P Coyne
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - G Wortmann
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - R K Nielsen
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - K Schaecher
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - C A Holland
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - U Krzych
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | | | - L A Soisson
- Malaria Vaccine Development Program, United States Agency for International Development, Washington, DC, USA
| | - E Angov
- Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| | - D G Heppner
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| |
Collapse
|
7
|
Zhou X, Zhang Q, Chen JH, Dai JF, Kassegne K. Revisiting the antigen markers of vector-borne parasitic diseases identified by immunomics: identification and application to disease control. Expert Rev Proteomics 2024; 21:205-216. [PMID: 38584506 DOI: 10.1080/14789450.2024.2336994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 03/03/2024] [Indexed: 04/09/2024]
Abstract
INTRODUCTION Protein microarray is a promising immunomic approach for identifying biomarkers. Based on our previous study that reviewed parasite antigens and recent parasitic omics research, this article expands to include information on vector-borne parasitic diseases (VBPDs), namely, malaria, schistosomiasis, leishmaniasis, babesiosis, trypanosomiasis, lymphatic filariasis, and onchocerciasis. AREAS COVERED We revisit and systematically summarize antigen markers of vector-borne parasites identified by the immunomic approach and discuss the latest advances in identifying antigens for the rational development of diagnostics and vaccines. The applications and challenges of this approach for VBPD control are also discussed. EXPERT OPINION The immunomic approach has enabled the identification and/or validation of antigen markers for vaccine development, diagnosis, disease surveillance, and treatment. However, this approach presents several challenges, including limited sample size, variability in antigen expression, false-positive results, complexity of omics data, validation and reproducibility, and heterogeneity of diseases. In addition, antigen involvement in host immune evasion and antigen sensitivity/specificity are major issues in its application. Despite these limitations, this approach remains promising for controlling VBPD. Advances in technology and data analysis methods should continue to improve candidate antigen identification, as well as the use of a multiantigen approach in diagnostic and vaccine development for VBPD control.
Collapse
Affiliation(s)
- Xia Zhou
- MOE Key Laboratory of Geriatric Diseases and Immunology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
| | - Qianqian Zhang
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Jun-Hu Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Parasitic Diseases, Chinese Center for Diseases Control and Prevention (Chinese Center for Tropical Diseases Research); National Health Commission of the People's Republic of China (NHC) Key Laboratory of Parasite and Vector Biology; World Health Organization (WHO) Collaborating Center for Tropical Diseases; National Center for International Research on Tropical Diseases, Shanghai, People's Republic of China
- Hainan Tropical Diseases Research Center (Hainan Sub-Center, Chinese Center for Tropical Diseases Research), Haikou, China
| | - Jian-Feng Dai
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Kokouvi Kassegne
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- One Health Center, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
8
|
Faber BW, Yeoh LM, Kurtovic L, Mol WEM, Poelert M, Smits E, Rodriguez Garcia R, Mandalawi-Van der Eijk M, van der Werff N, Voorberg-van der Wel A, Remarque EJ, Beeson JG, Kocken CHM. A Diversity Covering (DiCo) Plasmodium vivax apical membrane antigen-1 vaccine adjuvanted with RFASE/RSL10 yields high levels of growth-inhibitory antibodies. Vaccine 2024; 42:1785-1792. [PMID: 38365484 DOI: 10.1016/j.vaccine.2024.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 01/10/2024] [Accepted: 02/07/2024] [Indexed: 02/18/2024]
Abstract
Plasmodium vivax malaria is increasingly recognized as a major global health problem and the socio-economic impact of P.vivax-induced burden is huge. Vaccine development against P. vivax malaria has been hampered by the lack of an in vitro culture system and poor access to P. vivax sporozoites. The recent generation of Plasmodium falciparum parasites that express a functional P. vivax AMA1 molecule has provided a platform for in vitro evaluation of PvAMA1 as a potential blood stage vaccine. Three so-called PvAMA1 Diversity Covering (DiCo) proteins were designed to assess their potential to induce a functional and broad humoral immune response to the polymorphic PvAMA1 molecule. Rabbits were immunized with the mixture of three, Pichia-produced, PvAMA1 DiCo proteins, as well as with 2 naturally occurring PvAMA1 alleles. For these three groups, the experimental adjuvant raffinose fatty acid sulfate ester (RFASE) was used, while in a fourth group the purified main mono-esterified constituent (RSL10) of this adjuvant was used. Animals immunized with the mixture of the three PvAMA1 DiCo proteins in RFASE showed high anti-PvAMA1 antibody titers against three naturally occurring PvAMA1variants while also high growth-inhibitory capacity was observed against P. falciparum parasites expressing PvAMA1. This supports further clinical development of the PvAMA1 DiCo mixture as a potential malaria vaccine. However, as the single allele PvAMA1 SalI-group showed similar characteristics in antibody titer and inhibition levels as the PvAMA1 DiCo mixture-group, this raises the question whether a mixture is really necessary to overcome the polymorphism in the vaccine candidate. RFASE induced strong humoral responses, as did the animals immunized with the purified component, RSL10. This suggests that RSL10 is the active ingredient. However, one of the RSL10-immunized animal showed a delayed response, necessitating further research into the clinical development of RSL10.
Collapse
Affiliation(s)
- Bart W Faber
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, the Netherlands.
| | - Lee M Yeoh
- Burnet Institute, Melbourne, Victoria, Australia; Department of Medicine, The University of Melbourne, Victoria, Australia
| | - Liriye Kurtovic
- Burnet Institute, Melbourne, Victoria, Australia; Central Clinical School and Department of Microbiology, Monash University, Victoria, Australia
| | | | | | | | | | | | - Nicole van der Werff
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, the Netherlands
| | | | - Edmond J Remarque
- Department of Virology, Biomedical Primate Research Centre, Rijswijk, the Netherlands
| | - James G Beeson
- Burnet Institute, Melbourne, Victoria, Australia; Central Clinical School and Department of Microbiology, Monash University, Victoria, Australia; Department of Infectious Diseases, University of Melbourne, Victoria, Australia
| | - Clemens H M Kocken
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, the Netherlands
| |
Collapse
|
9
|
Hawadak J, Kojom Foko LP, Dongang Nana RR, Yadav K, Pande V, Das A, Singh V. Genetic diversity and natural selection of apical membrane antigen-1 (ama-1) in Cameroonian Plasmodium falciparum isolates. Gene 2024; 894:147956. [PMID: 37925116 DOI: 10.1016/j.gene.2023.147956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 10/16/2023] [Accepted: 10/31/2023] [Indexed: 11/06/2023]
Abstract
Antigenic variation associated with genetic diversity in global Plasmodium falciparum apical membrane antigen-1 (PfAMA-1) is a major impediment to designing an effective malaria vaccine. Here, we report the first study on genetic diversity and natural selection of the Pfama-1 gene in P. falciparum isolates from Cameroon. A total of 328 P. falciparum positive samples collected during 2016 and 2019 from five localities of Cameroon were analysed. The ectodomain coding fragment of Pfama-1 gene was amplified for polymorphism profiling and natural selection analysis. A total of 108 distinct haplotypes were found in 203 P. falciparum isolates with considerable nucleotide diversity (π = 0.016) and haplotype diversity (Hd = 0.976). Most amino acid substitutions detected were scattered in ectodomain-I and few specific mutations viz P145L, K148Q, K462I, L463F, N471K, S482L, E537G, K546R and I547F were seen only in Cameroonian isolates. A tendency of natural selection towards positive diversifying selection was observed (Taj-D = 2.058). Five positively selected codon sites (P145L, S283L, Q308E/K, P330S and I547F) were identified, which overlapped with predicted B-cell epitopes and red blood cell (RBC) binding sites, suggesting their potential implication in host immune pressure and parasite-RBC binding complex modulation. The Cameroonian P. falciparum populations indicated a moderate level of genetic differentiation when compared with global sequences, with few exceptions from Vietnam and Venezuela. Our findings provide baseline data on existing Pfama-1 gene polymorphisms in Cameroonian field isolates, which will be useful information for malaria vaccine design.
Collapse
Affiliation(s)
- Joseph Hawadak
- ICMR-National Institute of Malaria Research (NIMR), Delhi, India; Department of Biotechnology, Kumaun University, Bhimtal, Uttarakhand, India
| | - Loick Pradel Kojom Foko
- ICMR-National Institute of Malaria Research (NIMR), Delhi, India; Department of Biotechnology, Kumaun University, Bhimtal, Uttarakhand, India
| | - Rodrigue Roman Dongang Nana
- ICMR-National Institute of Malaria Research (NIMR), Delhi, India; Institut de Recherches Médicales et D'Etudes des Plantes Médicinales (IMPM), Yaoundé, Cameroon
| | - Karmveer Yadav
- ICMR-National Institute of Malaria Research (NIMR), Delhi, India
| | - Veena Pande
- Department of Biotechnology, Kumaun University, Bhimtal, Uttarakhand, India
| | - Aparup Das
- ICMR-National Institute for Research in Tribal Health (NIRTH), Jabalpur, India.
| | - Vineeta Singh
- ICMR-National Institute of Malaria Research (NIMR), Delhi, India.
| |
Collapse
|
10
|
Martinez FJ, White M, Guillotte-Blisnick M, Huon C, Boucharlat A, Agou F, England P, Popovici J, Hou MM, Silk SE, Barrett JR, Nielsen CM, Reimer JM, Mukherjee P, Chauhan VS, Minassian AM, Draper SJ, Chitnis CE. PvDBPII elicits multiple antibody-mediated mechanisms that reduce growth in a Plasmodium vivax challenge trial. NPJ Vaccines 2024; 9:10. [PMID: 38184681 PMCID: PMC10771494 DOI: 10.1038/s41541-023-00796-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/07/2023] [Indexed: 01/08/2024] Open
Abstract
The receptor-binding domain, region II, of the Plasmodium vivax Duffy binding protein (PvDBPII) binds the Duffy antigen on the reticulocyte surface to mediate invasion. A heterologous vaccine challenge trial recently showed that a delayed dosing regimen with recombinant PvDBPII SalI variant formulated with adjuvant Matrix-MTM reduced the in vivo parasite multiplication rate (PMR) in immunized volunteers challenged with the Thai P. vivax isolate PvW1. Here, we describe extensive analysis of the polyfunctional antibody responses elicited by PvDBPII immunization and identify immune correlates for PMR reduction. A classification algorithm identified antibody features that significantly contribute to PMR reduction. These included antibody titre, receptor-binding inhibitory titre, dissociation constant of the PvDBPII-antibody interaction, complement C1q and Fc gamma receptor binding and specific IgG subclasses. These data suggest that multiple immune mechanisms elicited by PvDBPII immunization are likely to be associated with protection and the immune correlates identified could guide the development of an effective vaccine for P. vivax malaria. Importantly, all the polyfunctional antibody features that correlated with protection cross-reacted with both PvDBPII SalI and PvW1 variants, suggesting that immunization with PvDBPII should protect against diverse P. vivax isolates.
Collapse
Affiliation(s)
- Francisco J Martinez
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, Paris, France
| | - Michael White
- Infectious Disease Epidemiology and Analytics G5 Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | | | - Christèle Huon
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, Paris, France
| | - Alix Boucharlat
- Chemogenomic and Biological Screening Core Facility, C2RT, Institut Pasteur, Université Paris Cité, CNRS UMR 3523, Paris, France
| | - Fabrice Agou
- Chemogenomic and Biological Screening Core Facility, C2RT, Institut Pasteur, Université Paris Cité, CNRS UMR 3523, Paris, France
| | - Patrick England
- Molecular Biophysics Facility, CNRS UMR 3528, Institut Pasteur, Paris, France
| | - Jean Popovici
- Malaria Reasearch Unit, Institut Pasteur du Cambodge, Pnom Penh, Cambodia
| | - Mimi M Hou
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK
| | - Sarah E Silk
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK
| | - Jordan R Barrett
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK
| | - Carolyn M Nielsen
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK
| | | | | | - Virander S Chauhan
- International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Angela M Minassian
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Simon J Draper
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Chetan E Chitnis
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, Paris, France.
| |
Collapse
|
11
|
Kibwana E, Kapulu M. Controlled Human Malaria Infection Studies in Africa-Past, Present, and Future. Curr Top Microbiol Immunol 2024; 445:337-365. [PMID: 35704094 PMCID: PMC7616462 DOI: 10.1007/82_2022_256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Controlled human infection studies have contributed significantly to the understanding of pathogeneses and treatment of infectious diseases. In malaria, deliberately infecting humans with malaria parasites was used as a treatment for neurosyphilis in the early 1920s. More recently, controlled human malaria infection (CHMI) has become a valuable, cost-effective tool to fast-track the development and evaluation of new anti-malarial drugs and/or vaccines. CHMI studies have also been used to define host/parasite interactions and immunological correlates of protection. CHMI involves infecting a small number of healthy volunteers with malaria parasites, monitoring their parasitemia and providing anti-malarial treatment when a set threshold is reached. In this review we discuss the introduction, development, and challenges of modern-day Plasmodium falciparum CHMI studies conducted in Africa, and the impact of naturally acquired immunity on infectivity and vaccine efficacy. CHMIs have shown to be an invaluable tool particularly in accelerating malaria vaccine research. Although there are limitations of CHMI studies for estimating public health impacts and for regulatory purposes, their strength lies in proof-of-concept efficacy data at an early stage of development, providing a faster way to select vaccines for further development and providing valuable insights in understanding the mechanisms of immunity to malarial infection.
Collapse
Affiliation(s)
- Elizabeth Kibwana
- Bioscience Department, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya
| | - Melissa Kapulu
- Bioscience Department, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya
| |
Collapse
|
12
|
Abo YN, Jamrozik E, McCarthy JS, Roestenberg M, Steer AC, Osowicki J. Strategic and scientific contributions of human challenge trials for vaccine development: facts versus fantasy. THE LANCET. INFECTIOUS DISEASES 2023; 23:e533-e546. [PMID: 37573871 DOI: 10.1016/s1473-3099(23)00294-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 08/15/2023]
Abstract
The unprecedented speed of delivery of SARS-CoV-2 pandemic vaccines has redefined the limits for all vaccine development. Beyond the aspirational 100-day timeline for tomorrow's hypothetical pandemic vaccines, there is a sense of optimism that development of other high priority vaccines can be accelerated. Early in the COVID-19 pandemic, an intense and polarised academic and public discourse arose concerning the role of human challenge trials for vaccine development. A case was made for human challenge trials as a powerful tool to establish early proof-of-concept of vaccine efficacy in humans, inform vaccine down selection, and address crucial knowledge gaps regarding transmission, pathogenesis, and immune protection. We review the track record of human challenge trials contributing to the development of vaccines for 19 different pathogens and discuss relevant limitations, barriers, and pitfalls. This Review also highlights opportunities for efforts to broaden the scope and boost the effects of human challenge trials, to accelerate all vaccine development.
Collapse
Affiliation(s)
- Yara-Natalie Abo
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia; Infectious Diseases Unit, Department of General Medicine, Royal Children's Hospital Melbourne, Parkville, VIC, Australia.
| | - Euzebiusz Jamrozik
- Ethox and Pandemic Sciences Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UK; Monash-WHO Collaborating Centre for Bioethics, Monash University, Melbourne, VIC, Australia
| | - James S McCarthy
- Department of Infectious Diseases, The University of Melbourne, Parkville, VIC, Australia; Victorian Infectious Diseases Services, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Meta Roestenberg
- Controlled Human Infections Center, Leiden University Medical Center, Leiden, Netherlands
| | - Andrew C Steer
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia; Infectious Diseases Unit, Department of General Medicine, Royal Children's Hospital Melbourne, Parkville, VIC, Australia
| | - Joshua Osowicki
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia; Infectious Diseases Unit, Department of General Medicine, Royal Children's Hospital Melbourne, Parkville, VIC, Australia
| |
Collapse
|
13
|
Rodríguez-Obediente K, Yepes-Pérez Y, Benavides-Ortiz D, Díaz-Arévalo D, Reyes C, Arévalo-Pinzón G, Patarroyo MA. Invasion-inhibitory peptides chosen by natural selection analysis as an antimalarial strategy. Mol Immunol 2023; 163:86-103. [PMID: 37769577 DOI: 10.1016/j.molimm.2023.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 09/06/2023] [Accepted: 09/14/2023] [Indexed: 10/03/2023]
Abstract
Plasmodium vivax's biological complexity has restricted in vitro culture development for characterising antigens involved in erythrocyte invasion and their immunological relevance. The murine model is proposed as a suitable alternative in the search for therapeutic candidates since Plasmodium yoelii uses homologous proteins for its invasion. The AMA-1 protein is essential for parasite invasion of erythrocytes as it is considered an important target for infection control. This study has focused on functional PyAMA-1 peptides involved in host-pathogen interaction; the protein is located in regions under negative selection as determined by bioinformatics analysis. It was found that pyama1 has two highly conserved regions amongst species (>70%) under negative selection. Fourteen synthetic peptides spanning both conserved regions were evaluated; 5 PyAMA-1 peptides having high specific binding (HABP) to murine erythrocytes were identified. The parasite's invasion inhibition capability was analysed through in vitro assays, suggesting that peptides 42681 (43-ENTERSIKLINPWDKYMEKY-62), 42903 (206-RYSSNDANNENQPFSFTPEK-225) and 42904 (221-FTPEKIENYKDLSYLTKNLR-240) had greater than 50% inhibition profile and restricted P. yoelii intra-erythrocyte development. This work proposes that the screening of conserved HABPs under negative selective pressure might be good candidates for developing a synthetic anti-malarial vaccine since they share functionally-relevant characteristics, such as interspecies conservation, specific RBC binding profile, invasion and parasite development inhibition capability, and the predicted B-epitopes within were recognised by sera obtained from experimentally-infected mice.
Collapse
Affiliation(s)
- Kewin Rodríguez-Obediente
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia; MSc programme in Microbiology, Biotechnology Institute, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Yoelis Yepes-Pérez
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Daniel Benavides-Ortiz
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia; School of Health Sciences, Universidad Colegio Mayor de Cundinamarca, Bogotá, Colombia
| | - Diana Díaz-Arévalo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia; Animal Science Faculty, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Bogotá, Colombia
| | - César Reyes
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Gabriela Arévalo-Pinzón
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia; Microbiology Department, Science Faculty, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Manuel Alfonso Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia; Microbiology Department, Faculty of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia.
| |
Collapse
|
14
|
Abstract
Malaria is a mosquito-borne disease caused by protozoan parasites of the genus Plasmodium. Despite significant declines in malaria-attributable morbidity and mortality over the last two decades, it remains a major public health burden in many countries. This underscores the critical need for improved strategies to prevent, treat and control malaria if we are to ultimately progress towards the eradication of this disease. Ideally, this will include the development and deployment of a highly effective malaria vaccine that is able to induce long-lasting protective immunity. There are many malaria vaccine candidates in development, with more than a dozen of these in clinical development. RTS,S/AS01 (also known as Mosquirix) is the most advanced malaria vaccine and was shown to have modest efficacy against clinical malaria in phase III trials in 5- to 17-month-old infants. Following pilot implementation trials, the World Health Organisation has recommended it for use in Africa in young children who are most at risk of infection with P. falciparum, the deadliest of the human malaria parasites. It is well recognised that more effective malaria vaccines are needed. In this review, we discuss malaria vaccine candidates that have progressed into clinical evaluation and highlight the most advanced candidates: Sanaria's irradiated sporozoite vaccine (PfSPZ Vaccine), the chemoattenuated sporozoite vaccine (PfSPZ-CVac), RTS,S/AS01 and the novel malaria vaccine candidate, R21, which displayed promising, high-level efficacy in a recent small phase IIb trial in Africa.
Collapse
Affiliation(s)
- Danielle I Stanisic
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia.
| | - Michael F Good
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia.
| |
Collapse
|
15
|
Silk SE, Kalinga WF, Mtaka IM, Lilolime NS, Mpina M, Milando F, Ahmed S, Diouf A, Mkwepu F, Simon B, Athumani T, Rashid M, Mohammed L, Lweno O, Ali AM, Nyaulingo G, Mwalimu B, Mswata S, Mwamlima TG, Barrett JR, Wang LT, Themistocleous Y, King LDW, Hodgson SH, Payne RO, Nielsen CM, Lawrie AM, Nugent FL, Cho JS, Long CA, Miura K, Draper SJ, Minassian AM, Olotu AI. Superior antibody immunogenicity of a viral-vectored RH5 blood-stage malaria vaccine in Tanzanian infants as compared to adults. MED 2023; 4:668-686.e7. [PMID: 37572659 DOI: 10.1016/j.medj.2023.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/23/2023] [Accepted: 07/11/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND RH5 is a leading blood-stage candidate antigen for a Plasmodium falciparum vaccine; however, its safety and immunogenicity in malaria-endemic populations are unknown. METHODS A phase 1b, single-center, dose-escalation, age-de-escalation, double-blind, randomized, controlled trial was conducted in Bagamoyo, Tanzania (NCT03435874). Between 12th April and 25th October 2018, 63 healthy adults (18-35 years), young children (1-6 years), and infants (6-11 months) received a priming dose of viral-vectored ChAd63 RH5 or rabies control vaccine. Sixty participants were boosted with modified vaccinia virus Ankara (MVA) RH5 or rabies control vaccine 8 weeks later and completed 6 months of follow-up post priming. Primary outcomes were the number of solicited and unsolicited adverse events post vaccination and the number of serious adverse events over the study period. Secondary outcomes included measures of the anti-RH5 immune response. FINDINGS Vaccinations were well tolerated, with profiles comparable across groups. No serious adverse events were reported. Vaccination induced RH5-specific cellular and humoral responses. Higher anti-RH5 serum immunoglobulin G (IgG) responses were observed post boost in young children and infants compared to adults. Vaccine-induced antibodies showed growth inhibition activity (GIA) in vitro against P. falciparum blood-stage parasites; their highest levels were observed in infants. CONCLUSIONS The ChAd63-MVA RH5 vaccine shows acceptable safety and reactogenicity and encouraging immunogenicity in children and infants residing in a malaria-endemic area. The levels of functional GIA observed in RH5-vaccinated infants are the highest reported to date following human vaccination. These data support onward clinical development of RH5-based blood-stage vaccines to protect against clinical malaria in young African infants. FUNDING Medical Research Council, London, UK.
Collapse
Affiliation(s)
- Sarah E Silk
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Wilmina F Kalinga
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Ivanny M Mtaka
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Nasoro S Lilolime
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Maximillian Mpina
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Florence Milando
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Saumu Ahmed
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Fatuma Mkwepu
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Beatus Simon
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Thabit Athumani
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Mohammed Rashid
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Latipha Mohammed
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Omary Lweno
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Ali M Ali
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Gloria Nyaulingo
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Bakari Mwalimu
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Sarah Mswata
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Tunu G Mwamlima
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Jordan R Barrett
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Lawrence T Wang
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Yrene Themistocleous
- Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK
| | - Lloyd D W King
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Susanne H Hodgson
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Ruth O Payne
- Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK
| | - Carolyn M Nielsen
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Alison M Lawrie
- Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK
| | - Fay L Nugent
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK
| | - Jee-Sun Cho
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Carole A Long
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Simon J Draper
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Angela M Minassian
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Ally I Olotu
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| |
Collapse
|
16
|
Yanik S, Venkatesh V, Parker ML, Ramaswamy R, Diouf A, Sarkar D, Miura K, Long CA, Boulanger MJ, Srinivasan P. Structure guided mimicry of an essential P. falciparum receptor-ligand complex enhances cross neutralizing antibodies. Nat Commun 2023; 14:5879. [PMID: 37735574 PMCID: PMC10514071 DOI: 10.1038/s41467-023-41636-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023] Open
Abstract
Invasion of human erythrocytes by Plasmodium falciparum (Pf) merozoites relies on the interaction between two parasite proteins: apical membrane antigen 1 (AMA1) and rhoptry neck protein 2 (RON2). While antibodies to AMA1 provide limited protection against Pf in non-human primate malaria models, clinical trials using recombinant AMA1 alone (apoAMA1) yielded no protection due to insufficient functional antibodies. Immunization with AMA1 bound to RON2L, a 49-amino acid peptide from its ligand RON2, has shown superior protection by increasing the proportion of neutralizing antibodies. However, this approach relies on the formation of a complex in solution between the two vaccine components. To advance vaccine development, here we engineered chimeric antigens by replacing the AMA1 DII loop, displaced upon ligand binding, with RON2L. Structural analysis confirmed that the fusion chimera (Fusion-FD12) closely mimics the binary AMA1-RON2L complex. Immunization studies in female rats demonstrated that Fusion-FD12 immune sera, but not purified IgG, neutralized vaccine-type parasites more efficiently compared to apoAMA1, despite lower overall anti-AMA1 titers. Interestingly, Fusion-FD12 immunization enhanced antibodies targeting conserved epitopes on AMA1, leading to increased neutralization of non-vaccine type parasites. Identifying these cross-neutralizing antibody epitopes holds promise for developing an effective, strain-transcending malaria vaccine.
Collapse
Affiliation(s)
- Sean Yanik
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Varsha Venkatesh
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Michelle L Parker
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 3P6, Canada
| | - Raghavendran Ramaswamy
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 3P6, Canada
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Deepti Sarkar
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Martin J Boulanger
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 3P6, Canada
| | - Prakash Srinivasan
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA.
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA.
| |
Collapse
|
17
|
Patel PN, Dickey TH, Diouf A, Salinas ND, McAleese H, Ouahes T, Long CA, Miura K, Lambert LE, Tolia NH. Structure-based design of a strain transcending AMA1-RON2L malaria vaccine. Nat Commun 2023; 14:5345. [PMID: 37660103 PMCID: PMC10475129 DOI: 10.1038/s41467-023-40878-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/14/2023] [Indexed: 09/04/2023] Open
Abstract
Apical membrane antigen 1 (AMA1) is a key malaria vaccine candidate and target of neutralizing antibodies. AMA1 binds to a loop in rhoptry neck protein 2 (RON2L) to form the moving junction during parasite invasion of host cells, and this complex is conserved among apicomplexan parasites. AMA1-RON2L complex immunization achieves higher growth inhibitory activity than AMA1 alone and protects mice against Plasmodium yoelii challenge. Here, three single-component AMA1-RON2L immunogens were designed that retain the structure of the two-component AMA1-RON2L complex: one structure-based design (SBD1) and two insertion fusions. All immunogens elicited high antibody titers with potent growth inhibitory activity, yet these antibodies did not block RON2L binding to AMA1. The SBD1 immunogen induced significantly more potent strain-transcending neutralizing antibody responses against diverse strains of Plasmodium falciparum than AMA1 or AMA1-RON2L complex vaccination. This indicates that SBD1 directs neutralizing antibody responses to strain-transcending epitopes in AMA1 that are independent of RON2L binding. This work underscores the importance of neutralization mechanisms that are distinct from RON2 blockade. The stable single-component SBD1 immunogen elicits potent strain-transcending protection that may drive the development of next-generation vaccines for improved malaria and apicomplexan parasite control.
Collapse
Affiliation(s)
- Palak N Patel
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Thayne H Dickey
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Nichole D Salinas
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Holly McAleese
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tarik Ouahes
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Lynn E Lambert
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Niraj H Tolia
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
18
|
Hou MM, Barrett JR, Themistocleous Y, Rawlinson TA, Diouf A, Martinez FJ, Nielsen CM, Lias AM, King LDW, Edwards NJ, Greenwood NM, Kingham L, Poulton ID, Khozoee B, Goh C, Hodgson SH, Mac Lochlainn DJ, Salkeld J, Guillotte-Blisnick M, Huon C, Mohring F, Reimer JM, Chauhan VS, Mukherjee P, Biswas S, Taylor IJ, Lawrie AM, Cho JS, Nugent FL, Long CA, Moon RW, Miura K, Silk SE, Chitnis CE, Minassian AM, Draper SJ. Vaccination with Plasmodium vivax Duffy-binding protein inhibits parasite growth during controlled human malaria infection. Sci Transl Med 2023; 15:eadf1782. [PMID: 37437014 PMCID: PMC7615121 DOI: 10.1126/scitranslmed.adf1782] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 06/05/2023] [Indexed: 07/14/2023]
Abstract
There are no licensed vaccines against Plasmodium vivax. We conducted two phase 1/2a clinical trials to assess two vaccines targeting P. vivax Duffy-binding protein region II (PvDBPII). Recombinant viral vaccines using chimpanzee adenovirus 63 (ChAd63) and modified vaccinia virus Ankara (MVA) vectors as well as a protein and adjuvant formulation (PvDBPII/Matrix-M) were tested in both a standard and a delayed dosing regimen. Volunteers underwent controlled human malaria infection (CHMI) after their last vaccination, alongside unvaccinated controls. Efficacy was assessed by comparisons of parasite multiplication rates in the blood. PvDBPII/Matrix-M, given in a delayed dosing regimen, elicited the highest antibody responses and reduced the mean parasite multiplication rate after CHMI by 51% (n = 6) compared with unvaccinated controls (n = 13), whereas no other vaccine or regimen affected parasite growth. Both viral-vectored and protein vaccines were well tolerated and elicited expected, short-lived adverse events. Together, these results support further clinical evaluation of the PvDBPII/Matrix-M P. vivax vaccine.
Collapse
Affiliation(s)
- Mimi M Hou
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Jordan R Barrett
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | | | | | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Francisco J Martinez
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Carolyn M Nielsen
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Amelia M Lias
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Lloyd D W King
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Nick J Edwards
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Lucy Kingham
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Ian D Poulton
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Cyndi Goh
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Susanne H Hodgson
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Dylan J Mac Lochlainn
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Jo Salkeld
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Micheline Guillotte-Blisnick
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Christèle Huon
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Franziska Mohring
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | | | - Virander S Chauhan
- International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | | | - Sumi Biswas
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Iona J Taylor
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Jee-Sun Cho
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Fay L Nugent
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Carole A Long
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Robert W Moon
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Sarah E Silk
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Chetan E Chitnis
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Angela M Minassian
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Simon J Draper
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
19
|
El-Moamly AA, El-Sweify MA. Malaria vaccines: the 60-year journey of hope and final success-lessons learned and future prospects. Trop Med Health 2023; 51:29. [PMID: 37198702 DOI: 10.1186/s41182-023-00516-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/18/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND The world has made great strides towards beating malaria, although about half of the world population is still exposed to the risk of contracting malaria. Developing an effective malaria vaccine was a huge challenge for medical science. In 2021 the World Health Organization (WHO) approved the first malaria vaccine, RTS,S/AS01 vaccine (Mosquirix™), for widespread use. This review highlights the history of development, and the different approaches and types of malaria vaccines, and the literature to date. It covers the developmental stages of RTS,S/AS01 and recommends steps for its deployment. The review explores other potential vaccine candidates and their status, and suggests options for their further development. It also recommends future roles for vaccines in eradicating malaria. Questions remain on how RTS,S vaccine will work in widespread use and how it can best be utilized to benefit vulnerable communities. CONCLUSION Malaria vaccines have been in development for almost 60 years. The RTS,S/AS01 vaccine has now been approved, but cannot be a stand-alone solution. Development should continue on promising candidates such as R21, PfSPZ and P. vivax vaccines. Multi-component vaccines may be a useful addition to other malaria control techniques in achieving eradication of malaria.
Collapse
Affiliation(s)
- Amal A El-Moamly
- Department of Medical Parasitology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Mohamed A El-Sweify
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
20
|
Srinivasan P, Yanik S, Venkatesh V, Parker M, Diouf A, Sarkar D, Miura K, Long C, Boulanger M. Structure guided mimicry of an essential P. falciparum receptor-ligand complex enhances cross neutralizing antibodies. RESEARCH SQUARE 2023:rs.3.rs-2733434. [PMID: 37131813 PMCID: PMC10153359 DOI: 10.21203/rs.3.rs-2733434/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Invasion of human red blood cells (RBCs) by Plasmodium falciparum (Pf) merozoites relies on the interaction between two parasite proteins, apical membrane antigen 1 (AMA1) and rhoptry neck protein 2 (RON2) 1,2 . Antibodies to AMA1 confer limited protection against P. falciparum in non-human primate malaria models 3,4 . However, clinical trials with recombinant AMA1 alone (apoAMA1) saw no protection, likely due to inadequate levels of functional antibodies 5-8 . Notably, immunization with AMA1 in its ligand bound conformation using RON2L, a 49 amino acid peptide from RON2, confers superior protection against P. falciparum malaria by enhancing the proportion of neutralizing antibodies 9,10 . A limitation of this approach, however, is that it requires the two vaccine components to form a complex in solution. To facilitate vaccine development, we engineered chimeric antigens by strategically replacing the AMA1 DII loop that is displaced upon ligand binding with RON2L. Structural characterization of the fusion chimera, Fusion-F D12 to 1.55 Å resolution showed that it closely mimics the binary receptor-ligand complex. Immunization studies showed that Fusion-F D12 immune sera neutralized parasites more efficiently than apoAMA1 immune sera despite having an overall lower anti-AMA1 titer, suggesting improvement in antibody quality. Furthermore, immunization with Fusion-F D12 enhanced antibodies targeting conserved epitopes on AMA1 resulting in greater neutralization of non-vaccine type parasites. Identifying epitopes of such cross-neutralizing antibodies will help in the development of an effective, strain-transcending malaria vaccine. Our fusion protein design is a robust vaccine platform that can be enhanced by incorporating polymorphisms in AMA1 to effectively neutralize all P. falciparum parasites.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Carole Long
- Laboratory of Malaria and Vector Resarch, NIAID/NIH
| | | |
Collapse
|
21
|
Roobsoong W, Yadava A, Draper SJ, Minassian AM, Sattabongkot J. The challenges of Plasmodium vivax human malaria infection models for vaccine development. Front Immunol 2023; 13:1006954. [PMID: 36685545 PMCID: PMC9849360 DOI: 10.3389/fimmu.2022.1006954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/09/2022] [Indexed: 01/07/2023] Open
Abstract
Controlled Human Malaria Infection models (CHMI) have been critical to advancing new vaccines for malaria. Stringent and safe preparation of a challenge agent is key to the success of any CHMI. Difficulty producing the Plasmodium vivax parasite in vitro has limited production of qualified parasites for CHMI as well as the functional assays required to screen and down-select candidate vaccines for this globally distributed parasite. This and other challenges to P. vivax CHMI (PvCHMI), including scientific, logistical, and ethical obstacles, are common to P. vivax research conducted in both non-endemic and endemic countries, with additional hurdles unique to each. The challenges of using CHMI for P. vivax vaccine development and evaluation, lessons learned from previous and ongoing clinical trials, and the way forward to effectively perform PvCHMI to support vaccine development, are discussed.
Collapse
Affiliation(s)
- Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Anjali Yadava
- Biologics Research & Development, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Simon J. Draper
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | | | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
22
|
Kim MJ, Chu KB, Lee SH, Kang HJ, Yoon KW, Ahmed MA, Quan FS. Recombinant Vaccinia Virus Expressing Plasmodium berghei Apical Membrane Antigen 1 or Microneme Protein Enhances Protection against P. berghei Infection in Mice. Trop Med Infect Dis 2022; 7:350. [PMID: 36355892 PMCID: PMC9698705 DOI: 10.3390/tropicalmed7110350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 07/27/2023] Open
Abstract
Recombinant vaccinia viruses (rVV) are effective antigen delivery vectors and are researched widely as vaccine platforms against numerous diseases. Apical membrane antigen 1 (AMA1) is one of the candidate antigens for malaria vaccines but rising concerns regarding its genetic diversity and polymorphism have necessitated the need to search for an alternative antigen. Here, we compare the efficacies of the rVV vaccines expressing either AMA1 or microneme protein (MIC) of Plasmodium berghei in mice. Mice (BALB/c) were immunized with either rVV-AMA1 or rVV-MIC and subsequently challenge-infected with P. berghei. Compared to the control group, both antigens elicited elevated levels of parasite-specific antibody responses. Immunization with either one of the two vaccines induced high levels of T cells and germinal center B cell responses. Interestingly, rVV-MIC immunization elicited higher levels of cellular immune response compared to rVV-AMA1 immunization, and significantly reduced pro-inflammatory cytokine productions were observed from the former vaccine. While differences in parasitemia and bodyweight changes were negligible between rVV-AMA1 and rVV-MIC immunization groups, prolonged survival was observed for the latter of the two. Based on these results, our findings suggest that the rVV expressing the P. berghei MIC could be a vaccine-candidate antigen.
Collapse
Affiliation(s)
- Min-Ju Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Ki-Back Chu
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Su-Hwa Lee
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Hae-Ji Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Keon-Woong Yoon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Md Atique Ahmed
- ICMR-Regional Medical Research Centre, NE Region, Dibrugarh 786010, Assam, India
| | - Fu-Shi Quan
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
23
|
Patarroyo ME, Bermudez A, Alba MP, Patarroyo MA, Suarez C, Aza-Conde J, Moreno-Vranich A, Vanegas M. Stereo electronic principles for selecting fully-protective, chemically-synthesised malaria vaccines. Front Immunol 2022; 13:926680. [DOI: 10.3389/fimmu.2022.926680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Major histocompatibility class II molecule-peptide-T-cell receptor (MHCII-p-TCR) complex-mediated antigen presentation for a minimal subunit-based, multi-epitope, multistage, chemically-synthesised antimalarial vaccine is essential for inducing an appropriate immune response. Deep understanding of this MHCII-p-TCR complex’s stereo-electronic characteristics is fundamental for vaccine development. This review encapsulates the main principles for achieving such epitopes’ perfect fit into MHC-II human (HLADRβ̞1*) or Aotus (Aona DR) molecules. The enormous relevance of several amino acids’ physico-chemical characteristics is analysed in-depth, as is data regarding a 26.5 ± 2.5Å distance between the farthest atoms fitting into HLA-DRβ1* structures’ Pockets 1 to 9, the role of polyproline II-like (PPIIL) structures having their O and N backbone atoms orientated for establishing H-bonds with specific HLA-DRβ1*-peptide binding region (PBR) residues. The importance of residues having specific charge and orientation towards the TCR for inducing appropriate immune activation, amino acids’ role and that of structures interfering with PPIIL formation and other principles are demonstrated which have to be taken into account when designing immune, protection-inducing peptide structures (IMPIPS) against diseases scourging humankind, malaria being one of them.
Collapse
|
24
|
Somanathan A, Mian SY, Chaddha K, Uchoi S, Bharti PK, Tandon R, Gaur D, Chauhan VS. Process development and preclinical evaluation of a major Plasmodium falciparum blood stage vaccine candidate, Cysteine-Rich Protective Antigen (CyRPA). Front Immunol 2022; 13:1005332. [PMID: 36211427 PMCID: PMC9535676 DOI: 10.3389/fimmu.2022.1005332] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
Plasmodium falciparum Cysteine-Rich Protective Antigen (CyRPA) is an essential, highly conserved merozoite antigen that forms an important multi-protein complex (RH5/Ripr/CyRPA) necessary for erythrocyte invasion. CyRPA is a promising blood-stage vaccine target that has been shown to elicit potent strain-transcending parasite neutralizing antibodies. Recently, we demonstrated that naturally acquired immune anti-CyRPA antibodies are invasion-inhibitory and therefore a correlate of protection against malaria. Here, we describe a process for the large-scale production of tag-free CyRPA vaccine in E. coli and demonstrate its parasite neutralizing efficacy with commonly used adjuvants. CyRPA was purified from inclusion bodies using a one-step purification method with high purity (>90%). Biochemical and biophysical characterization showed that the purified tag-free CyRPA interacted with RH5, readily detected by a conformation-specific CyRPA monoclonal antibody and recognized by sera from malaria infected individuals thus indicating that the recombinant antigen was correctly folded and retained its native conformation. Tag-free CyRPA formulated with Freund’s adjuvant elicited highly potent parasite neutralizing antibodies achieving inhibition of >90% across diverse parasite strains. Importantly, we identified tag-free CyRPA/Alhydrogel formulation as most effective in inducing a highly immunogenic antibody response that exhibited efficacious, cross-strain in vitro parasite neutralization achieving ~80% at 10 mg/ml. Further, CyRPA/Alhydrogel vaccine induced anti-parasite cytokine response in mice. In summary, our study provides a simple, scalable, cost-effective process for the production of tag-free CyRPA that in combination with human-compatible adjuvant induces efficacious humoral and cell-mediated immune response.
Collapse
Affiliation(s)
- Anjali Somanathan
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Syed Yusuf Mian
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Kritika Chaddha
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Seemalata Uchoi
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Praveen K. Bharti
- ICMR-National Institute of Research in Tribal Health (NIRTH), Jabalpur, India
| | - Ravi Tandon
- Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Deepak Gaur
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Virander Singh Chauhan
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
- *Correspondence: Virander Singh Chauhan,
| |
Collapse
|
25
|
Choy RKM, Bourgeois AL, Ockenhouse CF, Walker RI, Sheets RL, Flores J. Controlled Human Infection Models To Accelerate Vaccine Development. Clin Microbiol Rev 2022; 35:e0000821. [PMID: 35862754 PMCID: PMC9491212 DOI: 10.1128/cmr.00008-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The timelines for developing vaccines against infectious diseases are lengthy, and often vaccines that reach the stage of large phase 3 field trials fail to provide the desired level of protective efficacy. The application of controlled human challenge models of infection and disease at the appropriate stages of development could accelerate development of candidate vaccines and, in fact, has done so successfully in some limited cases. Human challenge models could potentially be used to gather critical information on pathogenesis, inform strain selection for vaccines, explore cross-protective immunity, identify immune correlates of protection and mechanisms of protection induced by infection or evoked by candidate vaccines, guide decisions on appropriate trial endpoints, and evaluate vaccine efficacy. We prepared this report to motivate fellow scientists to exploit the potential capacity of controlled human challenge experiments to advance vaccine development. In this review, we considered available challenge models for 17 infectious diseases in the context of the public health importance of each disease, the diversity and pathogenesis of the causative organisms, the vaccine candidates under development, and each model's capacity to evaluate them and identify correlates of protective immunity. Our broad assessment indicated that human challenge models have not yet reached their full potential to support the development of vaccines against infectious diseases. On the basis of our review, however, we believe that describing an ideal challenge model is possible, as is further developing existing and future challenge models.
Collapse
Affiliation(s)
- Robert K. M. Choy
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | - A. Louis Bourgeois
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Richard I. Walker
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Jorge Flores
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| |
Collapse
|
26
|
Salkeld J, Themistocleous Y, Barrett JR, Mitton CH, Rawlinson TA, Payne RO, Hou MM, Khozoee B, Edwards NJ, Nielsen CM, Sandoval DM, Bach FA, Nahrendorf W, Ramon RL, Baker M, Ramos-Lopez F, Folegatti PM, Quinkert D, Ellis KJ, Poulton ID, Lawrie AM, Cho JS, Nugent FL, Spence PJ, Silk SE, Draper SJ, Minassian AM. Repeat controlled human malaria infection of healthy UK adults with blood-stage Plasmodium falciparum: Safety and parasite growth dynamics. Front Immunol 2022; 13:984323. [PMID: 36072606 PMCID: PMC9444061 DOI: 10.3389/fimmu.2022.984323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
In endemic settings it is known that natural malaria immunity is gradually acquired following repeated exposures. Here we sought to assess whether similar acquisition of blood-stage malaria immunity would occur following repeated parasite exposure by controlled human malaria infection (CHMI). We report the findings of repeat homologous blood-stage Plasmodium falciparum (3D7 clone) CHMI studies VAC063C (ClinicalTrials.gov NCT03906474) and VAC063 (ClinicalTrials.gov NCT02927145). In total, 24 healthy, unvaccinated, malaria-naïve UK adult participants underwent primary CHMI followed by drug treatment. Ten of these then underwent secondary CHMI in the same manner, and then six of these underwent a final tertiary CHMI. As with primary CHMI, malaria symptoms were common following secondary and tertiary infection, however, most resolved within a few days of treatment and there were no long term sequelae or serious adverse events related to CHMI. Despite detectable induction and boosting of anti-merozoite serum IgG antibody responses following each round of CHMI, there was no clear evidence of anti-parasite immunity (manifest as reduced parasite growth in vivo) conferred by repeated challenge with the homologous parasite in the majority of volunteers. However, three volunteers showed some variation in parasite growth dynamics in vivo following repeat CHMI that were either modest or short-lived. We also observed no major differences in clinical symptoms or laboratory markers of infection across the primary, secondary and tertiary challenges. However, there was a trend to more severe pyrexia after primary CHMI and the absence of a detectable transaminitis post-treatment following secondary and tertiary infection. We hypothesize that this could represent the initial induction of clinical immunity. Repeat homologous blood-stage CHMI is thus safe and provides a model with the potential to further the understanding of naturally acquired immunity to blood-stage infection in a highly controlled setting.
Collapse
Affiliation(s)
- Jo Salkeld
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Jordan R. Barrett
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Celia H. Mitton
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Ruth O. Payne
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Mimi M. Hou
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Baktash Khozoee
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Nick J. Edwards
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Carolyn M. Nielsen
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Diana Muñoz Sandoval
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Florian A. Bach
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Wiebke Nahrendorf
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Megan Baker
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | - Doris Quinkert
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Ian D. Poulton
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Alison M. Lawrie
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Jee-Sun Cho
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Fay L. Nugent
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Philip J. Spence
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh, United Kingdom
| | - Sarah E. Silk
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Simon J. Draper
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Angela M. Minassian
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- *Correspondence: Angela M. Minassian,
| |
Collapse
|
27
|
Abstract
"The Primate Malarias" book has been a uniquely important resource for multiple generations of scientists, since its debut in 1971, and remains pertinent to the present day. Indeed, nonhuman primates (NHPs) have been instrumental for major breakthroughs in basic and pre-clinical research on malaria for over 50 years. Research involving NHPs have provided critical insights and data that have been essential for malaria research on many parasite species, drugs, vaccines, pathogenesis, and transmission, leading to improved clinical care and advancing research goals for malaria control, elimination, and eradication. Whilst most malaria scientists over the decades have been studying Plasmodium falciparum, with NHP infections, in clinical studies with humans, or using in vitro culture or rodent model systems, others have been dedicated to advancing research on Plasmodium vivax, as well as on phylogenetically related simian species, including Plasmodium cynomolgi, Plasmodium coatneyi, and Plasmodium knowlesi. In-depth study of these four phylogenetically related species over the years has spawned the design of NHP longitudinal infection strategies for gathering information about ongoing infections, which can be related to human infections. These Plasmodium-NHP infection model systems are reviewed here, with emphasis on modern systems biological approaches to studying longitudinal infections, pathogenesis, immunity, and vaccines. Recent discoveries capitalizing on NHP longitudinal infections include an advanced understanding of chronic infections, relapses, anaemia, and immune memory. With quickly emerging new technological advances, more in-depth research and mechanistic discoveries can be anticipated on these and additional critical topics, including hypnozoite biology, antigenic variation, gametocyte transmission, bone marrow dysfunction, and loss of uninfected RBCs. New strategies and insights published by the Malaria Host-Pathogen Interaction Center (MaHPIC) are recapped here along with a vision that stresses the importance of educating future experts well trained in utilizing NHP infection model systems for the pursuit of innovative, effective interventions against malaria.
Collapse
Affiliation(s)
- Mary R Galinski
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
- Emory Vaccine Center, Emory University, Atlanta, GA, USA.
- Emory National Primate Research Center (Yerkes National Primate Research Center), Emory University, Atlanta, GA, USA.
| |
Collapse
|
28
|
Bjerkan L, Visweswaran GRR, Gudjonsson A, Labbé GM, Quinkert D, Pattinson DJ, Spång HCL, Draper SJ, Bogen B, Braathen R. APC-Targeted DNA Vaccination Against Reticulocyte-Binding Protein Homolog 5 Induces Plasmodium falciparum-Specific Neutralizing Antibodies and T Cell Responses. Front Immunol 2021; 12:720550. [PMID: 34733274 PMCID: PMC8558525 DOI: 10.3389/fimmu.2021.720550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/30/2021] [Indexed: 11/20/2022] Open
Abstract
Targeted delivery of antigen to antigen presenting cells (APCs) is an efficient way to induce robust antigen-specific immune responses. Here, we present a novel DNA vaccine that targets the Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5), a leading blood-stage antigen of the human malaria pathogen, to APCs. The vaccine is designed as bivalent homodimers where each chain is composed of an amino-terminal single chain fragment variable (scFv) targeting unit specific for major histocompatibility complex class II (MHCII) expressed on APCs, and a carboxyl-terminal antigenic unit genetically linked by the dimerization unit. This vaccine format, named “Vaccibody”, has previously been successfully applied for antigens from other infectious diseases including influenza and HIV, as well as for tumor antigens. Recently, the crystal structure and key functional antibody epitopes for the truncated version of PfRH5 (PfRH5ΔNL) were characterized, suggesting PfRH5ΔNL to be a promising candidate for next-generation PfRH5 vaccine design. In this study, we explored the APC-targeting strategy for a PfRH5ΔNL-containing DNA vaccine. BALB/c mice immunized with the targeted vaccine induced higher PfRH5-specific IgG1 antibody responses than those vaccinated with a non-targeted vaccine or antigen alone. The APC-targeted vaccine also efficiently induced rapid IFN-γ and IL-4 T cell responses. Furthermore, the vaccine-induced PfRH5-specific IgG showed inhibition of growth of the P. falciparum 3D7 clone parasite in vitro. Finally, sera obtained after vaccination with this targeted vaccine competed for the same epitopes as PfRH5-specific mAbs from vaccinated humans. Robust humoral responses were also induced by a similar P. vivax Duffy-binding protein (PvDBP)-containing targeted DNA vaccine. Our data highlight a novel targeted vaccine platform for the development of vaccines against blood-stage malaria.
Collapse
Affiliation(s)
- Louise Bjerkan
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | - Arnar Gudjonsson
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | - Doris Quinkert
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Heidi C L Spång
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Simon J Draper
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Bjarne Bogen
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Ranveig Braathen
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
29
|
Minassian AM, Themistocleous Y, Silk SE, Barrett JR, Kemp A, Quinkert D, Nielsen CM, Edwards NJ, Rawlinson TA, Ramos Lopez F, Roobsoong W, Ellis KJ, Cho JS, Aunin E, Otto TD, Reid AJ, Bach FA, Labbé GM, Poulton ID, Marini A, Zaric M, Mulatier M, Lopez Ramon R, Baker M, Mitton CH, Sousa JC, Rachaphaew N, Kumpitak C, Maneechai N, Suansomjit C, Piteekan T, Hou MM, Khozoee B, McHugh K, Roberts DJ, Lawrie AM, Blagborough AM, Nugent FL, Taylor IJ, Johnson KJ, Spence PJ, Sattabongkot J, Biswas S, Rayner JC, Draper SJ. Controlled human malaria infection with a clone of Plasmodium vivax with high quality genome assembly. JCI Insight 2021; 6:152465. [PMID: 34609964 PMCID: PMC8675201 DOI: 10.1172/jci.insight.152465] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Controlled human malaria infection (CHMI) provides a highly informative means to investigate host-pathogen interactions and enable in vivo proof-of-concept efficacy testing of new drugs and vaccines. However, unlike Plasmodium falciparum, well-characterized P. vivax parasites that are safe and suitable for use in modern CHMI models are limited. Here, two healthy malaria-naïve UK adults with universal donor blood group were safely infected with a clone of P. vivax from Thailand by mosquito-bite CHMI. Parasitemia developed in both volunteers and, prior to treatment, each volunteer donated blood to produce a cryopreserved stabilate of infected red blood cells. Following stringent safety screening, the parasite stabilate from one of these donors ("PvW1") was thawed and used to inoculate six healthy malaria-naïve UK adults by blood-stage CHMI, at three different dilutions. Parasitemia developed in all volunteers, who were then successfully drug treated. PvW1 parasite DNA was isolated and sequenced to produce a high quality genome assembly by using a hybrid assembly method. We analysed leading vaccine candidate antigens and multigene families, including the Vivax interspersed repeat (VIR) genes of which we identified 1145 in the PvW1 genome. Our genomic analysis will guide future assessment of candidate vaccines and drugs, as well as experimental medicine studies.
Collapse
Affiliation(s)
| | | | - Sarah E Silk
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Jordan R Barrett
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Alison Kemp
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Doris Quinkert
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Nick J Edwards
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | - Jee-Sun Cho
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Eerik Aunin
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Thomas D Otto
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Adam J Reid
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Florian A Bach
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Ian D Poulton
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Arianna Marini
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Marija Zaric
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Margaux Mulatier
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Megan Baker
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Celia H Mitton
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Jason C Sousa
- Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Maryland, United States of America
| | | | | | | | | | - Tianrat Piteekan
- Mahidol Vivax Research Unit, Mahidol University, Bangkok, Thailand
| | - Mimi M Hou
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Baktash Khozoee
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Kirsty McHugh
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - David J Roberts
- Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, United Kingdom
| | - Alison M Lawrie
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Fay L Nugent
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Iona J Taylor
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Philip J Spence
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Sumi Biswas
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Julian C Rayner
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
30
|
Ofori EA, Tetteh JKA, Frimpong A, Ganeshan H, Belmonte M, Peters B, Villasante E, Sedegah M, Ofori MF, Kusi KA. Comparison of the impact of allelic polymorphisms in PfAMA1 on the induction of T Cell responses in high and low malaria endemic communities in Ghana. Malar J 2021; 20:367. [PMID: 34507582 PMCID: PMC8431259 DOI: 10.1186/s12936-021-03900-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/28/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Malaria eradication requires a combined effort involving all available control tools, and these efforts would be complemented by an effective vaccine. The antigen targets of immune responses may show polymorphisms that can undermine their recognition by immune effectors and hence render vaccines based on antigens from a single parasite variant ineffective against other variants. This study compared the influence of allelic polymorphisms in Plasmodium falciparum apical membrane antigen 1 (PfAMA1) peptide sequences from three strains of P. falciparum (3D7, 7G8 and FVO) on their function as immunodominant targets of T cell responses in high and low malaria transmission communities in Ghana. METHODS Peripheral blood mononuclear cells (PBMCs) from 10 subjects from a high transmission area (Obom) and 10 subjects from a low transmission area (Legon) were tested against 15 predicted CD8 + T cell minimal epitopes within the PfAMA1 antigen of multiple parasite strains using IFN-γ ELISpot assay. The peptides were also tested in similar assays against CD8 + enriched PBMC fractions from the same subjects in an effort to characterize the responding T cell subsets. RESULTS In assays using unfractionated PBMCs, two subjects from the high transmission area, Obom, responded positively to four (26.7%) of the 15 tested peptides. None of the Legon subject PBMCs yielded positive peptide responses using unfractionated PBMCs. In assays with CD8 + enriched PBMCs, three subjects from Obom made positive recall responses to six (40%) of the 15 tested peptides, while only one subject from Legon made a positive recall response to a single peptide. Overall, 5 of the 20 study subjects who had positive peptide-specific IFN-γ recall responses were from the high transmission area, Obom. Furthermore, while subjects from Obom responded to peptides in PfAMA1 from multiple parasite strains, one subject from Legon responded to a peptide from 3D7 strain only. CONCLUSIONS The current data demonstrate the possibility of a real effect of PfAMA1 polymorphisms on the induction of T cell responses in malaria exposed subjects, and this effect may be more pronounced in communities with higher parasite exposure.
Collapse
Affiliation(s)
- Ebenezer A Ofori
- West Africa Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana.,Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - John K A Tetteh
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Augustina Frimpong
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Harini Ganeshan
- Malaria Department, Naval Medical Research Center, Silver Spring, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Maria Belmonte
- Malaria Department, Naval Medical Research Center, Silver Spring, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Bjoern Peters
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Eileen Villasante
- Malaria Department, Naval Medical Research Center, Silver Spring, USA
| | - Martha Sedegah
- Malaria Department, Naval Medical Research Center, Silver Spring, USA
| | - Michael F Ofori
- West Africa Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana.,Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Kwadwo A Kusi
- West Africa Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana. .,Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana.
| |
Collapse
|
31
|
Aguttu C, Okech BA, Mukisa A, Lubega GW. Screening and characterization of hypothetical proteins of Plasmodium falciparum as novel vaccine candidates in the fight against malaria using reverse vaccinology. J Genet Eng Biotechnol 2021; 19:103. [PMID: 34269931 PMCID: PMC8283385 DOI: 10.1186/s43141-021-00199-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/16/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Plasmodium falciparum is the most deadly and leading cause of morbidity and mortality in Africa. About 90% of all malaria deaths in the world today occur in Sub-Saharan Africa especially in children aged < 5 years. In 2018, it was reported that there were 228 million malaria cases that resulted in 405,000 deaths from 91 countries. Currently, a fully effective and long-lasting preventive malaria vaccine is still elusive therefore more effort is needed to identify better effective vaccine candidates. The aim of this study was to identify and characterize hypothetical proteins as vaccine candidates derived from Plasmodium falciparum 3D7 genome by reverse vaccinology. RESULTS Of the 23 selected hypothetical proteins, 5 were predicted on the extracellular localization by WoLFPSORTv.2.0 program and all the 5 had less than 2 transmembrane regions that were predicted by TMHMMv2.0 and HMMTOP programs at default settings. Two out of the five proteins lacked secretory signal peptides as predicted by SignalP program. Among the 5 extracellular proteins, 3 were predicted to be antigenic by VaxiJen (score ≥ 0.5) and had negative GRAVY values ranging from - 1.156 to - 0.440. B cell epitope prediction by ABCpred and BCpred programs revealed a total of 15 antigenic epitopes. A total of 13 cytotoxic T cells were predicted from the 3 proteins using CTLPred online server. Only 2 out of the 13 CTL were antigenic, immunogenic, non-allergenic, and non-toxic using VaxiJen, IEDB, AllergenFp, and Toxinpred servers respectively in that order. Five HTL peptides from XP_001351030.1 protein are predicted inducers of all the three cytokines. STRING protein-protein network analysis of HPs revealed XP_001350955.1 closely interacts with nucleoside diphosphate kinase (PF13-0349) at 0.704, XP_001351030.1 interacts with male development protein1 (Mdv-1) at 0.645, and XP_001351047.1 with an uncharacterized protein (MAL8P1.53) at 0.400. CONCLUSION Reverse vaccinology is a promising strategy for the screening and identification of antigenic antigens with potential capacity to elicit cellular and humoral immune responses against P. falciparum infection. In this study, potential vaccine candidates of Plasmodium falciparum were identified and screened using standard bioinformatics tools. The vaccine candidates contained antigenic and immunogenic epitopes which could be considered for novel and effective vaccine targets. However, we strongly recommend in vivo and in vitro experiments to validate their immunogenicity and protective efficacy to completely decipher the vaccine targets against malaria.
Collapse
Affiliation(s)
- Claire Aguttu
- Department of Biochemistry and Sports Science, College of Natural Sciences, Makerere University, P.O. Box 7062, Kampala, Uganda
| | | | - Ambrose Mukisa
- Department of Biochemistry and Sports Science, College of Natural Sciences, Makerere University, P.O. Box 7062, Kampala, Uganda
| | - George William Lubega
- Department of Bio-molecular Resources and Bio-lab Sciences, School of Biosecurity, Biotechnology and Laboratory Sciences (SBLS), College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere University, P.O Box 7062, Kampala, Uganda
| |
Collapse
|
32
|
Minassian AM, Silk SE, Barrett JR, Nielsen CM, Miura K, Diouf A, Loos C, Fallon JK, Michell AR, White MT, Edwards NJ, Poulton ID, Mitton CH, Payne RO, Marks M, Maxwell-Scott H, Querol-Rubiera A, Bisnauthsing K, Batra R, Ogrina T, Brendish NJ, Themistocleous Y, Rawlinson TA, Ellis KJ, Quinkert D, Baker M, Lopez Ramon R, Ramos Lopez F, Barfod L, Folegatti PM, Silman D, Datoo M, Taylor IJ, Jin J, Pulido D, Douglas AD, de Jongh WA, Smith R, Berrie E, Noe AR, Diggs CL, Soisson LA, Ashfield R, Faust SN, Goodman AL, Lawrie AM, Nugent FL, Alter G, Long CA, Draper SJ. Reduced blood-stage malaria growth and immune correlates in humans following RH5 vaccination. MED 2021; 2:701-719.e19. [PMID: 34223402 PMCID: PMC8240500 DOI: 10.1016/j.medj.2021.03.014] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/19/2021] [Accepted: 03/25/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Development of an effective vaccine against the pathogenic blood-stage infection of human malaria has proved challenging, and no candidate vaccine has affected blood-stage parasitemia following controlled human malaria infection (CHMI) with blood-stage Plasmodium falciparum. METHODS We undertook a phase I/IIa clinical trial in healthy adults in the United Kingdom of the RH5.1 recombinant protein vaccine, targeting the P. falciparum reticulocyte-binding protein homolog 5 (RH5), formulated in AS01B adjuvant. We assessed safety, immunogenicity, and efficacy against blood-stage CHMI. Trial registered at ClinicalTrials.gov, NCT02927145. FINDINGS The RH5.1/AS01B formulation was administered using a range of RH5.1 protein vaccine doses (2, 10, and 50 μg) and was found to be safe and well tolerated. A regimen using a delayed and fractional third dose, in contrast to three doses given at monthly intervals, led to significantly improved antibody response longevity over ∼2 years of follow-up. Following primary and secondary CHMI of vaccinees with blood-stage P. falciparum, a significant reduction in parasite growth rate was observed, defining a milestone for the blood-stage malaria vaccine field. We show that growth inhibition activity measured in vitro using purified immunoglobulin G (IgG) antibody strongly correlates with in vivo reduction of the parasite growth rate and also identify other antibody feature sets by systems serology, including the plasma anti-RH5 IgA1 response, that are associated with challenge outcome. CONCLUSIONS Our data provide a new framework to guide rational design and delivery of next-generation vaccines to protect against malaria disease. FUNDING This study was supported by USAID, UK MRC, Wellcome Trust, NIAID, and the NIHR Oxford-BRC.
Collapse
Affiliation(s)
| | - Sarah E. Silk
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | | | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Carolin Loos
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Ashlin R. Michell
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Michael T. White
- Department of Parasites and Insect Vectors, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Nick J. Edwards
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Ian D. Poulton
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Celia H. Mitton
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Ruth O. Payne
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Michael Marks
- Centre for Clinical Infection and Diagnostics Research, King’s College London and Guy’s & St Thomas’ NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK
| | - Hector Maxwell-Scott
- Centre for Clinical Infection and Diagnostics Research, King’s College London and Guy’s & St Thomas’ NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK
| | - Antonio Querol-Rubiera
- Centre for Clinical Infection and Diagnostics Research, King’s College London and Guy’s & St Thomas’ NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK
| | - Karen Bisnauthsing
- Centre for Clinical Infection and Diagnostics Research, King’s College London and Guy’s & St Thomas’ NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK
| | - Rahul Batra
- Centre for Clinical Infection and Diagnostics Research, King’s College London and Guy’s & St Thomas’ NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK
| | - Tatiana Ogrina
- NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Nathan J. Brendish
- NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | | | | | | | - Doris Quinkert
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Megan Baker
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | | | - Lea Barfod
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Daniel Silman
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Mehreen Datoo
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Iona J. Taylor
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Jing Jin
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - David Pulido
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Willem A. de Jongh
- ExpreSion Biotechnologies, SCION-DTU Science Park, Agern Allé 1, Hørsholm 2970, Denmark
| | - Robert Smith
- Clinical BioManufacturing Facility, University of Oxford, Oxford OX3 7JT, UK
| | - Eleanor Berrie
- Clinical BioManufacturing Facility, University of Oxford, Oxford OX3 7JT, UK
| | | | | | | | | | - Saul N. Faust
- NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Anna L. Goodman
- Centre for Clinical Infection and Diagnostics Research, King’s College London and Guy’s & St Thomas’ NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK
| | | | - Fay L. Nugent
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Galit Alter
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Carole A. Long
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Simon J. Draper
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|
33
|
Accelerated phase Ia/b evaluation of the malaria vaccine candidate PfAMA1 DiCo demonstrates broadening of humoral immune responses. NPJ Vaccines 2021; 6:55. [PMID: 33854065 PMCID: PMC8046791 DOI: 10.1038/s41541-021-00319-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 03/17/2021] [Indexed: 11/08/2022] Open
Abstract
Plasmodium falciparum apical membrane antigen 1 (PfAMA1) is a candidate malaria vaccine antigen expressed on merozoites and sporozoites. PfAMA1's polymorphic nature impacts vaccine-induced protection. To address polymorphism, three Diversity Covering (DiCo) protein sequences were designed and tested in a staggered phase Ia/b trial. A cohort of malaria-naive adults received PfAMA1-DiCo adjuvanted with Alhydrogel® or GLA-SE and a cohort of malaria-exposed adults received placebo or GLA-SE adjuvanted PfAMA1 DiCo at weeks 0, 4 and 26. IgG and GIA levels measured 4 weeks after the third vaccination are similar in malaria-naive volunteers and placebo-immunised malaria-exposed adults, and have a similar breadth. Vaccination of malaria-exposed adults results in significant antibody level increases to the DiCo variants, but not to naturally occurring PfAMA1 variants. Moreover, GIA levels do not increase following vaccination. Future research will need to focus on stronger adjuvants and/or adapted vaccination regimens, to induce potentially protective responses in the target group of the vaccine.
Collapse
|
34
|
Pirahmadi S, Zakeri S, Djadid ND, Mehrizi AA. A review of combination adjuvants for malaria vaccines: a promising approach for vaccine development. Int J Parasitol 2021; 51:699-717. [PMID: 33798560 DOI: 10.1016/j.ijpara.2021.01.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/18/2020] [Accepted: 01/28/2021] [Indexed: 01/16/2023]
Abstract
It is obvious that there is a critical need for an efficient malaria vaccine to accelerate malaria eradication. Currently, recombinant subunit vaccination against malaria using proteins and peptides is gaining attention. However, one of the major drawbacks of this approach is the lack of an efficient and durable immune response. Therefore, subunit vaccines require adjuvants to make the vaccine sufficiently immunogenic. Considering the history of the RTS,S vaccine, it seems likely that no single adjuvant is capable of eliciting all the protective immune responses required in many malarial subunit vaccines and the use of combination adjuvants will be increasingly important as the science of malaria vaccines advances. In light of this, it appears that identifying the most effective mixture of adjuvants with minimal adverse effects offers tremendous opportunities in improving the efficacy of vaccines against malaria. Owing to the importance of a multi-adjuvanted approach in subunit malaria vaccine development, this review paper outlines some of the best known combination adjuvants used in malaria subunit vaccines, focusing on their proposed mechanisms of action, their immunological properties, and their notable results. The aim of the present review is to consolidate these findings to aid the application of these combination adjuvants in experimental malaria vaccines.
Collapse
Affiliation(s)
- Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Navid D Djadid
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Akram A Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
35
|
Arora N, C Anbalagan L, Pannu AK. Towards Eradication of Malaria: Is the WHO's RTS,S/AS01 Vaccination Effective Enough? Risk Manag Healthc Policy 2021; 14:1033-1039. [PMID: 33737844 PMCID: PMC7966294 DOI: 10.2147/rmhp.s219294] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/26/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Recent advances in mosquito eradication and antimalarial treatments have reduced the malaria burden only modestly. An effective malaria vaccine remains a high priority, but its development has several challenges. Among many potential candidates, the RTS,S/AS01 vaccine (MosquirixTM) remains the leading candidate. OBJECTIVE AND METHOD This review aims to understand the advances in the RTS,S/AS01 vaccine, and future comments regarding the vaccine's effectiveness in malaria eradication. Literature review for the past five decades was performed searching PubMed, EMBASE Ovid, and Cochrane Library, with using the following search items: ("malaria" OR "WHO's malaria" OR "Plasmodium falciparum" OR "RTS,S" OR "RTS,S/AS01" OR "RTS,S/AS02" OR "pre-erythrocytic malaria" OR "circumsporozoite" OR "Mosquirix") AND ("vaccine" OR "vaccination"). RESULTS RTS,S/AS01, a recombinant pre-erythrocytic vaccine containing Plasmodium falciparum surface-protein (circumsporozoite) antigen, is safe, well-tolerated, and immunogenic in children. Three doses, along with a booster, have a modest efficacy of about 36% in children (age 5-17 months) and about 26% in infants (age 6-12 weeks) against clinical malaria during a 48-month follow-up. However, the efficacy varies among population subgroups and with the parasite strain, it reduces without a booster and offers protection for a limited duration. Because of its potential cost-effectiveness and positive public health effect, the vaccine is being investigated in a pilot program for mortality benefits and broader deployment. CONCLUSION The RTS,S/AS01 vaccine prevents malaria; however, it should be considered another addition to the malaria-control program and not as an eradication tool because of its relatively low to modest efficacy.
Collapse
Affiliation(s)
- Navneet Arora
- Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Ashok K Pannu
- Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
36
|
Abstract
Introduction: An effective vaccine against malaria forms a global health priority. Both naturally acquired immunity and sterile protection induced by irradiated sporozoite immunization were described decades ago. Still no vaccine exists that sufficiently protects children in endemic areas. Identifying immunological correlates of vaccine efficacy can inform rational vaccine design and potentially accelerate clinical development.Areas covered: We discuss recent research on immunological correlates of malaria vaccine efficacy, including: insights from state-of-the-art omics platforms and systems vaccinology analyses; functional anti-parasitic assays; pre-immunization predictors of vaccine efficacy; and comparison of correlates of vaccine efficacy against controlled human malaria infections (CHMI) and against naturally acquired infections.Expert Opinion: Effective vaccination may be achievable without necessarily understanding immunological correlates, but the relatively disappointing efficacy of malaria vaccine candidates in target populations is concerning. Hypothesis-generating omics and systems vaccinology analyses, alongside assessment of pre-immunization correlates, have the potential to bring about paradigm-shifts in malaria vaccinology. Functional assays may represent in vivo effector mechanisms, but have scarcely been formally assessed as correlates. Crucially, evidence is still meager that correlates of vaccine efficacy against CHMI correspond with those against naturally acquired infections in target populations. Finally, the diversity of immunological assays and efficacy endpoints across malaria vaccine trials remains a major confounder.
Collapse
Affiliation(s)
| | - Matthew B B McCall
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, The Netherlands.,Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| |
Collapse
|
37
|
Milne K, Ivens A, Reid AJ, Lotkowska ME, O'Toole A, Sankaranarayanan G, Munoz Sandoval D, Nahrendorf W, Regnault C, Edwards NJ, Silk SE, Payne RO, Minassian AM, Venkatraman N, Sanders MJ, Hill AVS, Barrett M, Berriman M, Draper SJ, Rowe JA, Spence PJ. Mapping immune variation and var gene switching in naive hosts infected with Plasmodium falciparum. eLife 2021; 10:e62800. [PMID: 33648633 PMCID: PMC7924948 DOI: 10.7554/elife.62800] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
Falciparum malaria is clinically heterogeneous and the relative contribution of parasite and host in shaping disease severity remains unclear. We explored the interaction between inflammation and parasite variant surface antigen (VSA) expression, asking whether this relationship underpins the variation observed in controlled human malaria infection (CHMI). We uncovered marked heterogeneity in the host response to blood challenge; some volunteers remained quiescent, others triggered interferon-stimulated inflammation and some showed transcriptional evidence of myeloid cell suppression. Significantly, only inflammatory volunteers experienced hallmark symptoms of malaria. When we tracked temporal changes in parasite VSA expression to ask whether variants associated with severe disease rapidly expand in naive hosts, we found no transcriptional evidence to support this hypothesis. These data indicate that parasite variants that dominate severe malaria do not have an intrinsic growth or survival advantage; instead, they presumably rely upon infection-induced changes in their within-host environment for selection.
Collapse
Affiliation(s)
- Kathryn Milne
- Institute of Immunology and Infection Research, University of EdinburghEdinburghUnited Kingdom
| | - Alasdair Ivens
- Institute of Immunology and Infection Research, University of EdinburghEdinburghUnited Kingdom
- Centre for Immunity, Infection and Evolution, University of EdinburghEdinburghUnited Kingdom
| | - Adam J Reid
- Wellcome Sanger InstituteCambridgeUnited Kingdom
| | | | - Aine O'Toole
- Centre for Immunity, Infection and Evolution, University of EdinburghEdinburghUnited Kingdom
- Institute of Evolutionary Biology, University of EdinburghEdinburghUnited Kingdom
| | | | - Diana Munoz Sandoval
- Institute of Immunology and Infection Research, University of EdinburghEdinburghUnited Kingdom
- Instituto de Microbiologia, Universidad San Francisco de QuitoQuitoEcuador
| | - Wiebke Nahrendorf
- Institute of Immunology and Infection Research, University of EdinburghEdinburghUnited Kingdom
| | - Clement Regnault
- Wellcome Centre for Integrative Parasitology, University of GlasgowGlasgowUnited Kingdom
- Glasgow Polyomics, University of GlasgowGlasgowUnited Kingdom
| | - Nick J Edwards
- The Jenner Institute, University of OxfordOxfordUnited Kingdom
| | - Sarah E Silk
- The Jenner Institute, University of OxfordOxfordUnited Kingdom
| | - Ruth O Payne
- The Jenner Institute, University of OxfordOxfordUnited Kingdom
| | | | | | | | - Adrian VS Hill
- The Jenner Institute, University of OxfordOxfordUnited Kingdom
| | - Michael Barrett
- Wellcome Centre for Integrative Parasitology, University of GlasgowGlasgowUnited Kingdom
- Glasgow Polyomics, University of GlasgowGlasgowUnited Kingdom
| | | | - Simon J Draper
- The Jenner Institute, University of OxfordOxfordUnited Kingdom
| | - J Alexandra Rowe
- Institute of Immunology and Infection Research, University of EdinburghEdinburghUnited Kingdom
- Centre for Immunity, Infection and Evolution, University of EdinburghEdinburghUnited Kingdom
| | - Philip J Spence
- Institute of Immunology and Infection Research, University of EdinburghEdinburghUnited Kingdom
- Centre for Immunity, Infection and Evolution, University of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
38
|
Wockner LF, Hoffmann I, Webb L, Mordmüller B, Murphy SC, Kublin JG, O'Rourke P, McCarthy JS, Marquart L. Growth Rate of Plasmodium falciparum: Analysis of Parasite Growth Data From Malaria Volunteer Infection Studies. J Infect Dis 2020; 221:963-972. [PMID: 31679015 PMCID: PMC7198127 DOI: 10.1093/infdis/jiz557] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 10/29/2019] [Indexed: 12/16/2022] Open
Abstract
Background Growth rate of malaria parasites in the blood of infected subjects is an important
measure of efficacy of drugs and vaccines. Methods We used log-linear and sine-wave models to estimate the parasite growth rate of the 3D7
strain of Plasmodium falciparum using data from 177 subjects from 14
induced blood stage malaria (IBSM) studies conducted at QIMR Berghofer. We estimated
parasite multiplication rate per 48 hour (PMR48), PMR per life-cycle
(PMRLC), and parasite life-cycle duration. We compared these parameters to
those from studies conducted elsewhere with infections induced by IBSM (n=66),
sporozoites via mosquito bite (n=336) or injection (n=51). Results The parasite growth rate of 3D7 in QIMR Berghofer studies was 0.75/day (95% CI:
0.73–0.77/day), PMR48 was 31.9 (95% CI: 28.7–35.4),
PMRLC was 16.4 (95% CI: 15.1–17.8) and parasite life-cycle was 38.8
hour (95% CI: 38.3–39.2 hour). These parameters were similar to estimates from
IBSM studies elsewhere (0.71/day, 95% CI: 0.67–0.75/day; PMR48 26.6,
95% CI: 22.2–31.8), but significantly higher (P < 0.001)
than in sporozoite studies (0.47/day, 95% CI: 0.43–0.50/day; PMR48
8.6, 95% CI: 7.3–10.1). Conclusions Parasite growth rates were similar across different IBSM studies and higher than
infections induced by sporozoite.
Collapse
Affiliation(s)
- Leesa F Wockner
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Isabell Hoffmann
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Lachlan Webb
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Sean C Murphy
- Departments of Laboratory Medicine and Microbiology, University of Washington, Seattle, Washington, USA
| | - James G Kublin
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Peter O'Rourke
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Louise Marquart
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
39
|
Diversify and Conquer: The Vaccine Escapism of Plasmodium falciparum. Microorganisms 2020; 8:microorganisms8111748. [PMID: 33171746 PMCID: PMC7694999 DOI: 10.3390/microorganisms8111748] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022] Open
Abstract
Over the last century, a great deal of effort and resources have been poured into the development of vaccines to protect against malaria, particularly targeting the most widely spread and deadly species of the human-infecting parasites: Plasmodium falciparum. Many of the known proteins the parasite uses to invade human cells have been tested as vaccine candidates. However, precisely because of the importance and immune visibility of these proteins, they tend to be very diverse, and in many cases redundant, which limits their efficacy in vaccine development. With the advent of genomics and constantly improving sequencing technologies, an increasingly clear picture is emerging of the vast genomic diversity of parasites from different geographic areas. This diversity is distributed throughout the genome and includes most of the vaccine candidates tested so far, playing an important role in the low efficacy achieved. Genomics is a powerful tool to search for genes that comply with the most desirable attributes of vaccine targets, allowing us to evaluate function, immunogenicity and also diversity in the worldwide parasite populations. Even predicting how this diversity might evolve and spread in the future becomes possible, and can inform novel vaccine efforts.
Collapse
|
40
|
Bliss' and Loewe's additive and synergistic effects in Plasmodium falciparum growth inhibition by AMA1-RON2L, RH5, RIPR and CyRPA antibody combinations. Sci Rep 2020; 10:11802. [PMID: 32678144 PMCID: PMC7366652 DOI: 10.1038/s41598-020-67877-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/15/2020] [Indexed: 01/10/2023] Open
Abstract
Plasmodium invasion of red blood cells involves malaria proteins, such as reticulocyte-binding protein homolog 5 (RH5), RH5 interacting protein (RIPR), cysteine-rich protective antigen (CyRPA), apical membrane antigen 1 (AMA1) and rhoptry neck protein 2 (RON2), all of which are blood-stage malaria vaccine candidates. So far, vaccines containing AMA1 alone have been unsuccessful in clinical trials. However, immunization with AMA1 bound with RON2L (AMA1-RON2L) induces better protection against P. falciparum malaria in Aotus monkeys. We therefore sought to determine whether combinations of RH5, RIPR, CyRPA and AMA1-RON2L antibodies improve their biological activities and sought to develop a robust method for determination of synergy or additivity in antibody combinations. Rabbit antibodies against AMA1-RON2L, RH5, RIPR or CyRPA were tested either alone or in combinations in P. falciparum growth inhibition assay to determine Bliss' and Loewe's additivities. The AMA1-RON2L/RH5 combination consistently demonstrated an additive effect while the CyRPA/RIPR combination showed a modest synergistic effect with Hewlett’s \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$S=1.07 \left[95\% \mathrm{C}\mathrm{I}: 1.03, 1.19\right].$$\end{document}S=1.0795%CI:1.03,1.19. Additionally, we provide a publicly-available, online tool to aid researchers in analyzing and planning their own synergy experiments. This study supports future blood-stage vaccine development by providing a solid methodology to evaluate additive and/or synergistic (or antagonistic) effect of vaccine-induced antibodies.
Collapse
|
41
|
Duffy PE, Patrick Gorres J. Malaria vaccines since 2000: progress, priorities, products. NPJ Vaccines 2020; 5:48. [PMID: 32566259 PMCID: PMC7283239 DOI: 10.1038/s41541-020-0196-3] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/14/2020] [Indexed: 02/07/2023] Open
Abstract
Malaria vaccine development entered a new era in 2015 when the pre-erythrocytic Plasmodium falciparum candidate RTS,S was favorably reviewed by the European Medicines Agency and subsequently introduced into national pilot implementation programs, marking the first human anti-parasite vaccine to pass regulatory scrutiny. Since the first trials published in 1997, RTS,S has been evaluated in a series of clinical trials culminating in Phase 3 testing, while testing of other pre-erythrocytic candidates (that target sporozoite- or liver-stage parasites), particularly whole sporozoite vaccines, has also increased. Interest in blood-stage candidates (that limit blood-stage parasite growth) subsided after disappointing human efficacy results, although new blood-stage targets and concepts may revive activity in this area. Over the past decade, testing of transmission-blocking vaccines (that kill mosquito/sexual-stage parasites) advanced to field trials and the first generation of placental malaria vaccines (that clear placenta-sequestering parasites) entered the clinic. Novel antigen discovery, human monoclonal antibodies, structural vaccinology, and improved platforms promise to expand on RTS,S and improve existing vaccine candidates.
Collapse
Affiliation(s)
- Patrick E. Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - J. Patrick Gorres
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|
42
|
Ragotte RJ, Higgins MK, Draper SJ. The RH5-CyRPA-Ripr Complex as a Malaria Vaccine Target. Trends Parasitol 2020; 36:545-559. [PMID: 32359873 PMCID: PMC7246332 DOI: 10.1016/j.pt.2020.04.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/03/2020] [Accepted: 04/03/2020] [Indexed: 11/04/2022]
Abstract
Despite ongoing efforts, a highly effective vaccine against Plasmodium falciparum remains elusive. Vaccines targeting the pre-erythrocytic stages of the P. falciparum life cycle are the most advanced to date, affording moderate levels of efficacy in field trials. However, the discovery that the members of the merozoite PfRH5-PfCyRPA-PfRipr (RCR) complex are capable of inducing strain-transcendent neutralizing antibodies has renewed enthusiasm for the possibility of preventing disease by targeting the parasite during the blood stage of infection. With Phase I/II clinical trials now underway using first-generation vaccines against PfRH5, and more on the horizon for PfCyRPA and PfRipr, this review explores the rationale and future potential of the RCR complex as a P. falciparum vaccine target.
Collapse
Affiliation(s)
- Robert J Ragotte
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK.
| | - Matthew K Higgins
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK.
| |
Collapse
|
43
|
de Jong RM, Tebeje SK, Meerstein‐Kessel L, Tadesse FG, Jore MM, Stone W, Bousema T. Immunity against sexual stage Plasmodium falciparum and Plasmodium vivax parasites. Immunol Rev 2020; 293:190-215. [PMID: 31840844 PMCID: PMC6973022 DOI: 10.1111/imr.12828] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/30/2019] [Accepted: 11/14/2019] [Indexed: 12/25/2022]
Abstract
The efficient spread of malaria from infected humans to mosquitoes is a major challenge for malaria elimination initiatives. Gametocytes are the only Plasmodium life stage infectious to mosquitoes. Here, we summarize evidence for naturally acquired anti-gametocyte immunity and the current state of transmission blocking vaccines (TBV). Although gametocytes are intra-erythrocytic when present in infected humans, developing Plasmodium falciparum gametocytes may express proteins on the surface of red blood cells that elicit immune responses in naturally exposed individuals. This immune response may reduce the burden of circulating gametocytes. For both P. falciparum and Plasmodium vivax, there is a solid evidence that antibodies against antigens present on the gametocyte surface, when co-ingested with gametocytes, can influence transmission to mosquitoes. Transmission reducing immunity, reducing the burden of infection in mosquitoes, is a well-acknowledged but poorly quantified phenomenon that forms the basis for the development of TBV. Transmission enhancing immunity, increasing the likelihood or intensity of transmission to mosquitoes, is more speculative in nature but is convincingly demonstrated for P. vivax. With the increased interest in malaria elimination, TBV and monoclonal antibodies have moved to the center stage of malaria vaccine development. Methodologies to prioritize and evaluate products are urgently needed.
Collapse
MESH Headings
- Antibodies, Blocking/immunology
- Antibodies, Protozoan/immunology
- Host-Parasite Interactions/immunology
- Humans
- Immunity
- Immunomodulation
- Life Cycle Stages
- Malaria Vaccines/immunology
- Malaria, Falciparum/immunology
- Malaria, Falciparum/parasitology
- Malaria, Falciparum/prevention & control
- Malaria, Falciparum/transmission
- Malaria, Vivax/immunology
- Malaria, Vivax/parasitology
- Malaria, Vivax/prevention & control
- Malaria, Vivax/transmission
- Plasmodium falciparum/growth & development
- Plasmodium falciparum/immunology
- Plasmodium vivax/growth & development
- Plasmodium vivax/immunology
Collapse
Affiliation(s)
- Roos M. de Jong
- Radboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | | | - Lisette Meerstein‐Kessel
- Radboud Institute for Health SciencesRadboud University Medical CenterNijmegenThe Netherlands
- Centre for Molecular and Biomolecular InformaticsRadboud Institute for Molecular Life SciencesNijmegenThe Netherlands
| | - Fitsum G. Tadesse
- Armauer Hansen Research InstituteAddis AbabaEthiopia
- Radboud Institute for Health SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Matthijs M. Jore
- Radboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Will Stone
- Department of Immunology and InfectionLondon School of Hygiene and Tropical MedicineLondonUK
| | - Teun Bousema
- Radboud Institute for Health SciencesRadboud University Medical CenterNijmegenThe Netherlands
- Department of Immunology and InfectionLondon School of Hygiene and Tropical MedicineLondonUK
| |
Collapse
|
44
|
Atmar RL, Keitel WA. Searching for Improved Flu Vaccines-The Time Is Now. J Infect Dis 2020; 221:1-4. [PMID: 31665360 DOI: 10.1093/infdis/jiz545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022] Open
Affiliation(s)
- Robert L Atmar
- Departments of Medicine and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Wendy A Keitel
- Departments of Medicine and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
45
|
Salamanca DR, Gómez M, Camargo A, Cuy-Chaparro L, Molina-Franky J, Reyes C, Patarroyo MA, Patarroyo ME. Plasmodium falciparum Blood Stage Antimalarial Vaccines: An Analysis of Ongoing Clinical Trials and New Perspectives Related to Synthetic Vaccines. Front Microbiol 2019; 10:2712. [PMID: 31849871 PMCID: PMC6901501 DOI: 10.3389/fmicb.2019.02712] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 11/08/2019] [Indexed: 01/10/2023] Open
Abstract
Plasmodium falciparum malaria is a disease causing high morbidity and mortality rates worldwide, mainly in sub-Saharan Africa. Candidates have been identified for vaccines targeting the parasite's blood stage; this stage is important in the development of symptoms and clinical complications. However, no vaccine that can directly affect morbidity and mortality rates is currently available. This review analyzes the formulation, methodological design, and results of active clinical trials for merozoite-stage vaccines, regarding their safety profile, immunological response (phase Ia/Ib), and protective efficacy levels (phase II). Most vaccine candidates are in phase I trials and have had an acceptable safety profile. GMZ2 has made the greatest progress in clinical trials; its efficacy has been 14% in children aged less than 5 years in a phase IIb trial. Most of the available candidates that have shown strong immunogenicity and that have been tested for their protective efficacy have provided good results when challenged with a homologous parasite strain; however, their efficacy has dropped when they have been exposed to a heterologous strain. In view of these vaccines' unpromising results, an alternative approach for selecting new candidates is needed; such line of work should be focused on how to increase an immune response induced against the highly conserved (i.e., common to all strains), functionally relevant, protein regions that the parasite uses to invade target cells. Despite binding regions tending to be conserved, they are usually poorly antigenic and/or immunogenic, being frequently discarded as vaccine candidates when the conventional immunological approach is followed. The Fundación Instituto de Inmunología de Colombia (FIDIC) has developed a logical and rational methodology based on including conserved high-activity binding peptides (cHABPs) from the main P. falciparum biologically functional proteins involved in red blood cell (RBC) invasion. Once appropriately modified (mHABPs), these minimal, subunit-based, chemically synthesized peptides can be used in a system covering the human immune system's main genetic variables (the human leukocyte antigen HLA-DR isotype) inducing a suitable, immunogenic, and protective immune response in most of the world's populations.
Collapse
Affiliation(s)
- David Ricardo Salamanca
- Fundación Instituto de Inmunología de Colombia, Bogotá, Colombia.,Ph.D. Programme in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia.,Medicine Programme, Health Sciences Faculty, Universidad de Boyacá, Tunja, Colombia
| | - Marcela Gómez
- Fundación Instituto de Inmunología de Colombia, Bogotá, Colombia.,Ph.D. Programme in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia.,Medicine Programme, Health Sciences Faculty, Universidad de Boyacá, Tunja, Colombia
| | - Anny Camargo
- Fundación Instituto de Inmunología de Colombia, Bogotá, Colombia.,Ph.D. Programme in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia.,Medicine Programme, Health Sciences Faculty, Universidad de Boyacá, Tunja, Colombia
| | - Laura Cuy-Chaparro
- Fundación Instituto de Inmunología de Colombia, Bogotá, Colombia.,Ph.D. Programme in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia.,Medicine Programme, Health Sciences Faculty, Universidad de Boyacá, Tunja, Colombia
| | - Jessica Molina-Franky
- Fundación Instituto de Inmunología de Colombia, Bogotá, Colombia.,Ph.D. Programme in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia.,Medicine Programme, Health Sciences Faculty, Universidad de Boyacá, Tunja, Colombia
| | - César Reyes
- Fundación Instituto de Inmunología de Colombia, Bogotá, Colombia.,Ph.D. Programme in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Manuel Alfonso Patarroyo
- Fundación Instituto de Inmunología de Colombia, Bogotá, Colombia.,Basic Sciences Department, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Manuel Elkin Patarroyo
- Fundación Instituto de Inmunología de Colombia, Bogotá, Colombia.,Department of Pathology, School of Medicine, Universidad Nacional de Colombia, Boyacá, Colombia
| |
Collapse
|
46
|
Dejon-Agobe JC, Ateba-Ngoa U, Lalremruata A, Homoet A, Engelhorn J, Nouatin OP, Edoa JR, Fernandes JF, Esen M, Mouwenda YD, Betouke Ongwe EM, Massinga-Loembe M, Hoffman SL, Sim BKL, Theisen M, Kremsner PG, Adegnika AA, Lell B, Mordmüller B. Controlled Human Malaria Infection of Healthy Adults With Lifelong Malaria Exposure to Assess Safety, Immunogenicity, and Efficacy of the Asexual Blood Stage Malaria Vaccine Candidate GMZ2. Clin Infect Dis 2019; 69:1377-1384. [PMID: 30561539 PMCID: PMC6763635 DOI: 10.1093/cid/ciy1087] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 12/18/2018] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND GMZ2 is a recombinant malaria vaccine inducing immune responses against Plasmodium falciparum (Pf) merozoite surface protein-3 and glutamate-rich protein. We used standardized controlled human malaria infection (CHMI) to assess the efficacy of this asexual blood-stage vaccine. METHODS We vaccinated 50 healthy, adult volunteers with lifelong exposure to Pf 3 times, at 4-week intervals, with 30 or 100 µg GMZ2 formulated in CAF01, a liposome-based adjuvant; 100 µg GMZ2, formulated in Alhydrogel; or a control vaccine (Verorab). Approximately 13 weeks after the last vaccination, 35/50 volunteers underwent CHMI by direct venous inoculation of 3200 Pf sporozoites (Sanaria® PfSPZ Challenge). RESULTS Adverse events were similarly distributed between GMZ2 and control vaccinees. Baseline-corrected anti-GMZ2 antibody concentrations 4 weeks after the last vaccination were higher in all 3 GMZ2-vaccinated arms, compared to the control group. All GMZ2 formulations induced similar antibody levels. CHMI resulted in 29/34 (85%) volunteers with Pf parasitemia and 15/34 (44%) with malaria (parasitemia and symptoms). The proportion of participants with malaria (2/5 control, 6/10 GMZ2-Alhydrogel, 2/8 30 µg GMZ2-CAF01, and 5/11 100 µg GMZ2-CAF01) and the time it took them to develop malaria were similar in all groups. Baseline, vaccine-specific antibody concentrations were associated with protection against malaria. CONCLUSIONS GMZ2 is well tolerated and immunogenic in lifelong-Pf-exposed adults from Gabon, with similar antibody responses regardless of formulation. CHMI showed no protective effect of prior vaccination with GMZ2, although baseline, vaccine-specific antibody concentrations were associated with protection. CHMI with the PfSPZ Challenge is a potent new tool to validate asexual, blood-stage malaria vaccines in Africa. CLINICAL TRIALS REGISTRATION Pan-African Clinical Trials: PACTR201503001038304.
Collapse
Affiliation(s)
- Jean Claude Dejon-Agobe
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Ulysse Ateba-Ngoa
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
- Department of Parasitology, Leiden University Medical Center, The Netherlands
| | - Albert Lalremruata
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Andreas Homoet
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Julie Engelhorn
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Odilon Paterne Nouatin
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Jean Ronald Edoa
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - José F Fernandes
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Meral Esen
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Yoanne Darelle Mouwenda
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Department of Parasitology, Leiden University Medical Center, The Netherlands
| | - Eunice M Betouke Ongwe
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Marguerite Massinga-Loembe
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | | | | | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Denmark
- Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Peter G Kremsner
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Ayôla A Adegnika
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
- Department of Parasitology, Leiden University Medical Center, The Netherlands
| | - Bertrand Lell
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Benjamin Mordmüller
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| |
Collapse
|
47
|
Labbé GM, Miura K, Silk SE, Gu W, Moon JE, Jin J, Payne RO, Fay MP, Dutta S, Long CA, Draper SJ. Harmonization study between three laboratories for expression of malaria vaccine clinical trial IgG antibody ELISA data in µg/mL. Malar J 2019; 18:300. [PMID: 31477111 PMCID: PMC6721210 DOI: 10.1186/s12936-019-2935-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 08/24/2019] [Indexed: 11/11/2022] Open
Abstract
Background The ability to report vaccine-induced IgG responses in terms of µg/mL, as opposed arbitrary units (AU), enables a more informed interpretation of the magnitude of the immune response, and better comparison between vaccines targeting different antigens. However, these interpretations rely on the accuracy of the methodology, which is used to generate ELISA data in µg/mL. In a previous clinical trial of a vaccine targeting the apical membrane antigen 1 (AMA1) from Plasmodium falciparum, three laboratories (Oxford, NIH and WRAIR) reported ELISA data in µg/mL that were correlated but not concordant. This current study sought to harmonize the methodology used to generate a conversion factor (CF) for ELISA analysis of human anti-AMA1 IgG responses across the three laboratories. Methods Purified IgG was distributed to the three laboratories and, following a set protocol provided by NIH, AMA1-specific human IgG was affinity purified. A new “harmonized CF” was generated by each laboratory using their in-house ELISA, and the original clinical trial ELISA data were re-analysed accordingly. Results Statistical analysis showed that the data remained highly correlated across all three laboratories, although only Oxford and NIH were able to harmonize their CF for ELISA and generate concordant data. Conclusions This study enabled two out of the three laboratories to harmonize their µg/mL readouts for the human anti-AMA1 IgG ELISA, but results reported from WRAIR are ~ twofold higher. Given the need to validate such information for each species and antigen of interest, it is important to bear in mind these likely differences when interpreting µg/mL ELISA data in the future.
Collapse
Affiliation(s)
- Geneviève M Labbé
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD, 20852, USA
| | - Sarah E Silk
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Wenjuan Gu
- Biostatistics Research Branch, NIAID/NIH, Rockville, MD, 20852, USA
| | - James E Moon
- Military Malaria Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Jing Jin
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Ruth O Payne
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Michael P Fay
- Biostatistics Research Branch, NIAID/NIH, Rockville, MD, 20852, USA
| | - Sheetij Dutta
- Military Malaria Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD, 20852, USA
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK.
| |
Collapse
|
48
|
Cooper MM, Loiseau C, McCarthy JS, Doolan DL. Human challenge models: tools to accelerate the development of malaria vaccines. Expert Rev Vaccines 2019; 18:241-251. [PMID: 30732492 DOI: 10.1080/14760584.2019.1580577] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Malaria challenge models, where healthy human volunteers are intentionally infected with Plasmodium species parasites under controlled conditions, can be undertaken in several well-defined ways. These challenge models enable evaluation of the kinetics of parasite growth and clearance, host-pathogen interactions and the host immune response. They can facilitate discovery of candidate diagnostic biomarkers and novel vaccine targets. As translational tools they can facilitate testing of candidate vaccines and drugs and evaluation of diagnostic tests. AREAS COVERED Until recently, malaria human challenge models have been limited to only a few Plasmodium falciparum strains and used exclusively in malaria-naïve volunteers in non-endemic regions. Several recent advances include the use of alternate P. falciparum strains and other species of Plasmodia, as well as strains attenuated by chemical, radiation or genetic modification, and the conduct of studies in pre-exposed individuals. Herein, we discuss how this diversification is enabling more thorough vaccine efficacy testing and informing rational vaccine development. EXPERT OPINION The ability to comprehensively evaluate vaccine efficacy in controlled settings will continue to accelerate the translation of candidate malaria vaccines to the clinic, and inform the development and optimisation of potential vaccines that would be effective against multiple strains in geographically and demographically diverse settings.
Collapse
Affiliation(s)
- Martha M Cooper
- a Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| | - Claire Loiseau
- a Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| | - James S McCarthy
- b Infectious Diseases Programme , QIMR Berghofer Medical Research Institute , Brisbane , Australia
| | - Denise L Doolan
- a Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| |
Collapse
|
49
|
Akter J, Khoury DS, Aogo R, Lansink LIM, SheelaNair A, Thomas BS, Laohamonthonkul P, Pernold CPS, Dixon MWA, Soon MSF, Fogg LG, Engel JA, Elliott T, Sebina I, James KR, Cromer D, Davenport MP, Haque A. Plasmodium-specific antibodies block in vivo parasite growth without clearing infected red blood cells. PLoS Pathog 2019; 15:e1007599. [PMID: 30811498 PMCID: PMC6411214 DOI: 10.1371/journal.ppat.1007599] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/11/2019] [Accepted: 01/28/2019] [Indexed: 01/01/2023] Open
Abstract
Plasmodium parasites invade and multiply inside red blood cells (RBC). Through a cycle of maturation, asexual replication, rupture and release of multiple infective merozoites, parasitised RBC (pRBC) can reach very high numbers in vivo, a process that correlates with disease severity in humans and experimental animals. Thus, controlling pRBC numbers can prevent or ameliorate malaria. In endemic regions, circulating parasite-specific antibodies associate with immunity to high parasitemia. Although in vitro assays reveal that protective antibodies could control pRBC via multiple mechanisms, in vivo assessment of antibody function remains challenging. Here, we employed two mouse models of antibody-mediated immunity to malaria, P. yoelii 17XNL and P. chabaudi chabaudi AS infection, to study infection-induced, parasite-specific antibody function in vivo. By tracking a single generation of pRBC, we tested the hypothesis that parasite-specific antibodies accelerate pRBC clearance. Though strongly protective against homologous re-challenge, parasite-specific IgG did not alter the rate of pRBC clearance, even in the presence of ongoing, systemic inflammation. Instead, antibodies prevented parasites progressing from one generation of RBC to the next. In vivo depletion studies using clodronate liposomes or cobra venom factor, suggested that optimal antibody function required splenic macrophages and dendritic cells, but not complement C3/C5-mediated killing. Finally, parasite-specific IgG bound poorly to the surface of pRBC, yet strongly to structures likely exposed by the rupture of mature schizonts. Thus, in our models of humoral immunity to malaria, infection-induced antibodies did not accelerate pRBC clearance, and instead co-operated with splenic phagocytes to block subsequent generations of pRBC. Malaria occurs when Plasmodium parasites replicate inside red blood cells, with the number of parasitised cells (pRBC) correlating with disease severity. Antibodies are highly effective at controlling pRBC numbers in the bloodstream, and yet we know very little about how they function in vivo. Human in vitro studies predict that antibodies may function in a number of ways, including via phagocytes or different complement mechanisms. However, to date it has been challenging to explore how antibodies might control parasite numbers in vivo. Here, we have used a unique method in mice, where clearance and replication of a single cohort of pRBC was closely tracked in the presence of protective antibodies. Surprisingly, antibodies played no role whatsoever in accelerating the removal of pRBC. Instead, antibodies were highly effective at preventing parasites from progressing from one generation of pRBC to the next. This process partly depended on host phagocytes. However, we found no role for complement-mediated direct killing. Together, our in vivo data suggest in mouse models that naturally-acquired antibodies do not clear pRBC, and instead prevent transition from one red blood cell to the next.
Collapse
Affiliation(s)
- Jasmin Akter
- QIMR Berghofer Medical Research Institute, Herston, Brisbane QLD, Australia
| | - David S. Khoury
- Infection Analytics Program, Kirby Institute, UNSW Australia, Kensington NSW, Australia
| | - Rosemary Aogo
- Infection Analytics Program, Kirby Institute, UNSW Australia, Kensington NSW, Australia
| | | | - Arya SheelaNair
- QIMR Berghofer Medical Research Institute, Herston, Brisbane QLD, Australia
| | - Bryce S. Thomas
- QIMR Berghofer Medical Research Institute, Herston, Brisbane QLD, Australia
| | | | | | - Matthew W. A. Dixon
- University of Melbourne, Department of Biochemistry and Molecular Biology, Melbourne, Victoria, Australia
| | - Megan S. F. Soon
- QIMR Berghofer Medical Research Institute, Herston, Brisbane QLD, Australia
| | - Lily G. Fogg
- QIMR Berghofer Medical Research Institute, Herston, Brisbane QLD, Australia
| | - Jessica A. Engel
- QIMR Berghofer Medical Research Institute, Herston, Brisbane QLD, Australia
| | - Trish Elliott
- QIMR Berghofer Medical Research Institute, Herston, Brisbane QLD, Australia
| | - Ismail Sebina
- QIMR Berghofer Medical Research Institute, Herston, Brisbane QLD, Australia
| | - Kylie R. James
- QIMR Berghofer Medical Research Institute, Herston, Brisbane QLD, Australia
| | - Deborah Cromer
- Infection Analytics Program, Kirby Institute, UNSW Australia, Kensington NSW, Australia
| | - Miles P. Davenport
- Infection Analytics Program, Kirby Institute, UNSW Australia, Kensington NSW, Australia
- * E-mail: (MPD); (AH)
| | - Ashraful Haque
- QIMR Berghofer Medical Research Institute, Herston, Brisbane QLD, Australia
- * E-mail: (MPD); (AH)
| |
Collapse
|
50
|
Abstract
The development of highly effective and durable vaccines against the human malaria parasites Plasmodium falciparum and P. vivax remains a key priority. Decades of endeavor have taught that achieving this goal will be challenging; however, recent innovation in malaria vaccine research and a diverse pipeline of novel vaccine candidates for clinical assessment provides optimism. With first-generation pre-erythrocytic vaccines aiming for licensure in the coming years, it is important to reflect on how next-generation approaches can improve on their success. Here we review the latest vaccine approaches that seek to prevent malaria infection, disease, and transmission and highlight some of the major underlying immunological and molecular mechanisms of protection. The synthesis of rational antigen selection, immunogen design, and immunization strategies to induce quantitatively and qualitatively improved immune effector mechanisms offers promise for achieving sustained high-level protection.
Collapse
|