1
|
Callejas-Hernández F, Nikulkova M, Adamski N, Yan G, Yewhalaw D, Carlton JM. Assembled genome of an Ethiopian Plasmodium vivax isolate generated using GridION long-read technology. Microbiol Resour Announc 2024; 13:e0059024. [PMID: 39283158 PMCID: PMC11465822 DOI: 10.1128/mra.00590-24] [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: 06/05/2024] [Accepted: 08/27/2024] [Indexed: 10/11/2024] Open
Abstract
Plasmodium vivax causes ~20% of malaria cases in Ethiopia. Here, we report a long-read genome assembly generated from an individual collected in 2022. P. vivax is genetically diverse across endemic regions; thus, the genome assembly of an African isolate is an important resource for the malaria research community.
Collapse
Affiliation(s)
- Francisco Callejas-Hernández
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Maria Nikulkova
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, New York, USA
| | - Nicole Adamski
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, New York, USA
| | - Guiyun Yan
- Program in Public Health, University of California, Irvine, California, USA
| | - Delenasaw Yewhalaw
- Tropical and Infectious Diseases Research Centre, Jimma University, Jimma, Ethiopia
| | - Jane M. Carlton
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, New York, USA
| |
Collapse
|
2
|
Mertens JE, Rigby CA, Bardelli M, Quinkert D, Hou MM, Diouf A, Silk SE, Chitnis CE, Minassian AM, Moon RW, Long CA, Draper SJ, Miura K. Evaluation of the precision of the Plasmodium knowlesi growth inhibition assay for Plasmodium vivax Duffy-binding protein-based malaria vaccine development. Vaccine 2024; 42:3621-3629. [PMID: 38704253 PMCID: PMC11128340 DOI: 10.1016/j.vaccine.2024.04.073] [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: 03/01/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024]
Abstract
Recent data indicate increasing disease burden and importance of Plasmodium vivax (Pv) malaria. A robust assay will be essential for blood-stage Pv vaccine development. Results of the in vitro growth inhibition assay (GIA) with transgenic P. knowlesi (Pk) parasites expressing the Pv Duffy-binding protein region II (PvDBPII) correlate with in vivo protection in the first PvDBPII controlled human malaria infection (CHMI) trials, making the PkGIA an ideal selection tool once the precision of the assay is defined. To determine the precision in percentage of inhibition in GIA (%GIA) and in GIA50 (antibody concentration that gave 50 %GIA), ten GIAs with transgenic Pk parasites were conducted with four different anti-PvDBPII human monoclonal antibodies (mAbs) at concentrations of 0.016 to 2 mg/mL, and three GIAs with eighty anti-PvDBPII human polyclonal antibodies (pAbs) at 10 mg/mL. A significant assay-to-assay variation was observed, and the analysis revealed a standard deviation (SD) of 13.1 in the mAb and 5.94 in the pAb dataset for %GIA, with a LogGIA50 SD of 0.299 (for mAbs). Moreover, the ninety-five percent confidence interval (95 %CI) for %GIA or GIA50 in repeat assays was calculated in this investigation. The error range determined in this study will help researchers to compare PkGIA results from different assays and studies appropriately, thus supporting the development of future blood-stage malaria vaccine candidates, specifically second-generation PvDBPII-based formulations.
Collapse
Affiliation(s)
- Jonas E Mertens
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, United Kingdom; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, United Kingdom; Institute for Infection Research and Vaccine Development (IIRVD), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Cassandra A Rigby
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, United Kingdom; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, United Kingdom
| | - Martino Bardelli
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, United Kingdom; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, United Kingdom
| | - Doris Quinkert
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, United Kingdom; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, United Kingdom
| | - Mimi M Hou
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, United Kingdom; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, United Kingdom
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville 20852, MD, United States
| | - Sarah E Silk
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, United Kingdom; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, United Kingdom
| | - 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, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, United Kingdom; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, United Kingdom; NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Robert W Moon
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville 20852, MD, United States
| | - Simon J Draper
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, United Kingdom; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, United Kingdom; NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom.
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville 20852, MD, United States.
| |
Collapse
|
3
|
Kattenberg JH, Monsieurs P, De Meyer J, De Meulenaere K, Sauve E, de Oliveira TC, Ferreira MU, Gamboa D, Rosanas‐Urgell A. Population genomic evidence of structured and connected Plasmodium vivax populations under host selection in Latin America. Ecol Evol 2024; 14:e11103. [PMID: 38529021 PMCID: PMC10961478 DOI: 10.1002/ece3.11103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 03/27/2024] Open
Abstract
Pathogen genomic epidemiology has the potential to provide a deep understanding of population dynamics, facilitating strategic planning of interventions, monitoring their impact, and enabling timely responses, and thereby supporting control and elimination efforts of parasitic tropical diseases. Plasmodium vivax, responsible for most malaria cases outside Africa, shows high genetic diversity at the population level, driven by factors like sub-patent infections, a hidden reservoir of hypnozoites, and early transmission to mosquitoes. While Latin America has made significant progress in controlling Plasmodium falciparum, it faces challenges with residual P. vivax. To characterize genetic diversity and population structure and dynamics, we have analyzed the largest collection of P. vivax genomes to date, including 1474 high-quality genomes from 31 countries across Asia, Africa, Oceania, and America. While P. vivax shows high genetic diversity globally, Latin American isolates form a distinctive population, which is further divided into sub-populations and occasional clonal pockets. Genetic diversity within the continent was associated with the intensity of transmission. Population differentiation exists between Central America and the North Coast of South America, vs. the Amazon Basin, with significant gene flow within the Amazon Basin, but limited connectivity between the Northwest Coast and the Amazon Basin. Shared genomic regions in these parasite populations indicate adaptive evolution, particularly in genes related to DNA replication, RNA processing, invasion, and motility - crucial for the parasite's survival in diverse environments. Understanding these population-level adaptations is crucial for effective control efforts, offering insights into potential mechanisms behind drug resistance, immune evasion, and transmission dynamics.
Collapse
Affiliation(s)
| | - Pieter Monsieurs
- Malariology UnitInstitute of Tropical Medicine AntwerpAntwerpBelgium
| | - Julie De Meyer
- Malariology UnitInstitute of Tropical Medicine AntwerpAntwerpBelgium
- Present address:
Integrated Molecular Plant physiology Research (IMPRES) and Plants and Ecosystems (PLECO), Department of BiologyUniversity of AntwerpAntwerpBelgium
| | | | - Erin Sauve
- Malariology UnitInstitute of Tropical Medicine AntwerpAntwerpBelgium
| | - Thaís C. de Oliveira
- Department of Parasitology, Institute of Biomedical SciencesUniversity of São PauloSão PauloBrazil
| | - Marcelo U. Ferreira
- Department of Parasitology, Institute of Biomedical SciencesUniversity of São PauloSão PauloBrazil
- Global Health and Tropical Medicine, Institute of Hygiene and Tropical MedicineNova University of LisbonLisbonPortugal
| | - Dionicia Gamboa
- Instituto de Medicina Tropical “Alexander von Humboldt”Universidad Peruana Cayetano HerediaLimaPeru
- Laboratorio de Malaria: Parásitos y Vectores, Laboratorios de Investigación y Desarrollo, Departamento de Ciencias Celulares y Moleculares, Facultad de Ciencias e IngenieríaUniversidad Peruana Cayetano HerediaLimaPeru
| | | |
Collapse
|
4
|
Barrett JR, Silk SE, Mkindi CG, Kwiatkowska KM, Hou MM, Lias AM, Kalinga WF, Mtaka IM, McHugh K, Bardelli M, Davies H, King LDW, Edwards NJ, Chauhan VS, Mukherjee P, Rwezaula S, Chitnis CE, Olotu AI, Minassian AM, Draper SJ, Nielsen CM. Analyses of human vaccine-specific circulating and bone marrow-resident B cell populations reveal benefit of delayed vaccine booster dosing with blood-stage malaria antigens. Front Immunol 2024; 14:1193079. [PMID: 38299155 PMCID: PMC10827869 DOI: 10.3389/fimmu.2023.1193079] [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: 03/24/2023] [Accepted: 12/11/2023] [Indexed: 02/02/2024] Open
Abstract
We have previously reported primary endpoints of a clinical trial testing two vaccine platforms for the delivery of Plasmodium vivax malaria DBPRII: viral vectors (ChAd63, MVA), and protein/adjuvant (PvDBPII with 50µg Matrix-M™ adjuvant). Delayed boosting was necessitated due to trial halts during the pandemic and provides an opportunity to investigate the impact of dosing regimens. Here, using flow cytometry - including agnostic definition of B cell populations with the clustering tool CITRUS - we report enhanced induction of DBPRII-specific plasma cell and memory B cell responses in protein/adjuvant versus viral vector vaccinees. Within protein/adjuvant groups, delayed boosting further improved B cell immunogenicity compared to a monthly boosting regimen. Consistent with this, delayed boosting also drove more durable anti-DBPRII serum IgG. In an independent vaccine clinical trial with the P. falciparum malaria RH5.1 protein/adjuvant (50µg Matrix-M™) vaccine candidate, we similarly observed enhanced circulating B cell responses in vaccinees receiving a delayed final booster. Notably, a higher frequency of vaccine-specific (putatively long-lived) plasma cells was detected in the bone marrow of these delayed boosting vaccinees by ELISPOT and correlated strongly with serum IgG. Finally, following controlled human malaria infection with P. vivax parasites in the DBPRII trial, in vivo growth inhibition was observed to correlate with DBPRII-specific B cell and serum IgG responses. In contrast, the CD4+ and CD8+ T cell responses were impacted by vaccine platform but not dosing regimen and did not correlate with in vivo growth inhibition in a challenge model. Taken together, our DBPRII and RH5 data suggest an opportunity for protein/adjuvant dosing regimen optimisation in the context of rational vaccine development against pathogens where protection is antibody-mediated.
Collapse
Affiliation(s)
- Jordan R. Barrett
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Sarah E. Silk
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | - Mimi M. Hou
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Amelia M. Lias
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | - Kirsty McHugh
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Martino Bardelli
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Hannah Davies
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Lloyd D. W. King
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Nick J. Edwards
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Virander S. Chauhan
- International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | | | | | - Chetan E. Chitnis
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, Paris, France
| | | | - Angela M. Minassian
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Simon J. Draper
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Carolyn M. Nielsen
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
5
|
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
|
6
|
Bach FA, Muñoz Sandoval D, Mazurczyk M, Themistocleous Y, Rawlinson TA, Harding AC, Kemp A, Silk SE, Barrett JR, Edwards NJ, Ivens A, Rayner JC, Minassian AM, Napolitani G, Draper SJ, Spence PJ. A systematic analysis of the human immune response to Plasmodium vivax. J Clin Invest 2023; 133:e152463. [PMID: 37616070 PMCID: PMC10575735 DOI: 10.1172/jci152463] [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: 06/17/2021] [Accepted: 08/22/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUNDThe biology of Plasmodium vivax is markedly different from that of P. falciparum; how this shapes the immune response to infection remains unclear. To address this shortfall, we inoculated human volunteers with a clonal field isolate of P. vivax and tracked their response through infection and convalescence.METHODSParticipants were injected intravenously with blood-stage parasites and infection dynamics were tracked in real time by quantitative PCR. Whole blood samples were used for high dimensional protein analysis, RNA sequencing, and cytometry by time of flight, and temporal changes in the host response to P. vivax were quantified by linear regression. Comparative analyses with P. falciparum were then undertaken using analogous data sets derived from prior controlled human malaria infection studies.RESULTSP. vivax rapidly induced a type I inflammatory response that coincided with hallmark features of clinical malaria. This acute-phase response shared remarkable overlap with that induced by P. falciparum but was significantly elevated (at RNA and protein levels), leading to an increased incidence of pyrexia. In contrast, T cell activation and terminal differentiation were significantly increased in volunteers infected with P. falciparum. Heterogeneous CD4+ T cells were found to dominate this adaptive response and phenotypic analysis revealed unexpected features normally associated with cytotoxicity and autoinflammatory disease.CONCLUSIONP. vivax triggers increased systemic interferon signaling (cf P. falciparum), which likely explains its reduced pyrogenic threshold. In contrast, P. falciparum drives T cell activation far in excess of P. vivax, which may partially explain why falciparum malaria more frequently causes severe disease.TRIAL REGISTRATIONClinicalTrials.gov NCT03797989.FUNDINGThe European Union's Horizon 2020 Research and Innovation programme, the Wellcome Trust, and the Royal Society.
Collapse
Affiliation(s)
- Florian A. Bach
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Diana Muñoz Sandoval
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
- Insitute of Microbiology, Universidad San Francisco de Quito, Quito, Ecuador
| | | | | | | | - Adam C. Harding
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Alison Kemp
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Sarah E. Silk
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Jordan R. Barrett
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Nick J. Edwards
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Alasdair Ivens
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Julian C. Rayner
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Angela M. Minassian
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Giorgio Napolitani
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, and
| | - Simon J. Draper
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Philip J. Spence
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
7
|
De Meulenaere K, Cuypers B, Gamboa D, Laukens K, Rosanas-Urgell A. A new Plasmodium vivax reference genome for South American isolates. BMC Genomics 2023; 24:606. [PMID: 37821878 PMCID: PMC10568799 DOI: 10.1186/s12864-023-09707-5] [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: 03/14/2023] [Accepted: 09/30/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND Plasmodium vivax is the second most important cause of human malaria worldwide, and accounts for the majority of malaria cases in South America. A high-quality reference genome exists for Papua Indonesia (PvP01) and Thailand (PvW1), but is lacking for South America. A reference genome specifically for South America would be beneficial though, as P. vivax is a genetically diverse parasite with geographical clustering. RESULTS This study presents a new high-quality assembly of a South American P. vivax isolate, referred to as PvPAM (P. vivax Peruvian AMazon). The genome was obtained from a low input patient sample from the Peruvian Amazon and sequenced using PacBio technology, resulting in a highly complete assembly with 6497 functional genes. Telomeric ends were present in 17 out of 28 chromosomal ends, and additional (sub)telomeric regions are present in 12 unassigned contigs. A comparison of multigene families between PvPAM and the PvP01 genome revealed remarkable variation in vir genes, and the presence of merozoite surface proteins (MSP) 3.6 and 3.7. Three dhfr and dhps drug resistance associated mutations are present in PvPAM, similar to those found in other Peruvian isolates. Mapping of publicly available South American whole genome sequencing (WGS) data to PvPAM resulted in significantly fewer variants and truncated reads compared to the use of PvP01 or PvW1 as reference genomes. To minimize the number of core genome variants in non-South American samples, PvW1 is most suited for Southeast Asian isolates, both PvPAM and PvW1 are suited for South Asian isolates, and PvPAM is recommended for African isolates. Interestingly, non-South American samples still contained the least subtelomeric variants when mapped to PvPAM, indicating high quality of the PvPAM subtelomeric regions. CONCLUSIONS Our findings show that the PvPAM reference genome more accurately represents South American P. vivax isolates in comparison to PvP01 and PvW1. In addition, PvPAM has a high level of completeness, and contains a similar number of annotated genes as PvP01 or PvW1. The PvPAM genome therefore will be a valuable resource to improve future genomic analyses on P. vivax isolates from the South American continent.
Collapse
Affiliation(s)
- Katlijn De Meulenaere
- Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium.
- Department of Computer Science, University of Antwerp, Antwerp, Belgium.
| | - Bart Cuypers
- Department of Computer Science, University of Antwerp, Antwerp, Belgium
| | - Dionicia Gamboa
- Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima, Peru
- Departamento de Ciencias Celulares y Moleculares, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Kris Laukens
- Department of Computer Science, University of Antwerp, Antwerp, Belgium
| | - Anna Rosanas-Urgell
- Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium.
| |
Collapse
|
8
|
Weiss GE, Ragotte RJ, Quinkert D, Lias AM, Dans MG, Boulet C, Looker O, Ventura OD, Williams BG, Crabb BS, Draper SJ, Gilson PR. The dual action of human antibodies specific to Plasmodium falciparum PfRH5 and PfCyRPA: Blocking invasion and inactivating extracellular merozoites. PLoS Pathog 2023; 19:e1011182. [PMID: 37713419 PMCID: PMC10529537 DOI: 10.1371/journal.ppat.1011182] [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] [Received: 02/02/2023] [Revised: 09/27/2023] [Accepted: 08/29/2023] [Indexed: 09/17/2023] Open
Abstract
The Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5) is the current leading blood-stage malaria vaccine candidate. PfRH5 functions as part of the pentameric PCRCR complex containing PTRAMP, CSS, PfCyRPA and PfRIPR, all of which are essential for infection of human red blood cells (RBCs). To trigger RBC invasion, PfRH5 engages with RBC protein basigin in a step termed the RH5-basigin binding stage. Although we know increasingly more about how antibodies specific for PfRH5 can block invasion, much less is known about how antibodies recognizing other members of the PCRCR complex can inhibit invasion. To address this, we performed live cell imaging using monoclonal antibodies (mAbs) which bind PfRH5 and PfCyRPA. We measured the degree and timing of the invasion inhibition, the stage at which it occurred, as well as subsequent events. We show that parasite invasion is blocked by individual mAbs, and the degree of inhibition is enhanced when combining a mAb specific for PfRH5 with one binding PfCyRPA. In addition to directly establishing the invasion-blocking capacity of the mAbs, we identified a secondary action of certain mAbs on extracellular parasites that had not yet invaded where the mAbs appeared to inactivate the parasites by triggering a developmental pathway normally only seen after successful invasion. These findings suggest that epitopes within the PfCyRPA-PfRH5 sub-complex that elicit these dual responses may be more effective immunogens than neighboring epitopes by both blocking parasites from invading and rapidly inactivating extracellular parasites. These two protective mechanisms, prevention of invasion and inactivation of uninvaded parasites, resulting from antibody to a single epitope indicate a possible route to the development of more effective vaccines.
Collapse
Affiliation(s)
- Greta E. Weiss
- Burnet Institute, 85 Commercial Road, Melbourne, Victoria, Australia
| | - Robert J. Ragotte
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, United Kingdom
| | - Doris Quinkert
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, United Kingdom
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, United Kingdom
| | - Amelia M. Lias
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, United Kingdom
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, United Kingdom
| | - Madeline G. Dans
- Burnet Institute, 85 Commercial Road, Melbourne, Victoria, Australia
| | - Coralie Boulet
- Burnet Institute, 85 Commercial Road, Melbourne, Victoria, Australia
| | - Oliver Looker
- Burnet Institute, 85 Commercial Road, Melbourne, Victoria, Australia
| | - Olivia D. Ventura
- Burnet Institute, 85 Commercial Road, Melbourne, Victoria, Australia
| | - Barnabas G. Williams
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, United Kingdom
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, United Kingdom
| | - Brendan S. Crabb
- Burnet Institute, 85 Commercial Road, Melbourne, Victoria, Australia
- The University of Melbourne, Grattan Street, Parkville, Victoria, Australia
| | - Simon J. Draper
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, United Kingdom
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, United Kingdom
| | - Paul R. Gilson
- Burnet Institute, 85 Commercial Road, Melbourne, Victoria, Australia
- The University of Melbourne, Grattan Street, Parkville, Victoria, Australia
| |
Collapse
|
9
|
Nicholas J, De SL, Thawornpan P, Brashear AM, Kolli SK, Subramani PA, Barnes SJ, Cui L, Chootong P, Ntumngia FB, Adams JH. Preliminary characterization of Plasmodium vivax sporozoite antigens as pre-erythrocytic vaccine candidates. PLoS Negl Trop Dis 2023; 17:e0011598. [PMID: 37703302 PMCID: PMC10519608 DOI: 10.1371/journal.pntd.0011598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/25/2023] [Accepted: 08/15/2023] [Indexed: 09/15/2023] Open
Abstract
Plasmodium vivax pre-erythrocytic (PE) vaccine research has lagged far behind efforts to develop Plasmodium falciparum vaccines. There is a critical gap in our knowledge of PE antigen targets that can induce functionally inhibitory neutralizing antibody responses. To overcome this gap and guide the selection of potential PE vaccine candidates, we considered key characteristics such as surface exposure, essentiality to infectivity and liver stage development, expression as recombinant proteins, and functional immunogenicity. Selected P. vivax sporozoite antigens were surface sporozoite protein 3 (SSP3), sporozoite microneme protein essential for cell traversal (SPECT1), sporozoite surface protein essential for liver-stage development (SPELD), and M2 domain of MAEBL. Sequence analysis revealed little variation occurred in putative B-cell and T-cell epitopes of the PE candidates. Each antigen was tested for expression as refolded recombinant proteins using an established bacterial expression platform and only SPELD failed. The successfully expressed antigens were immunogenic in vaccinated laboratory mice and were positively reactive with serum antibodies of P. vivax-exposed residents living in an endemic region in Thailand. Vaccine immune antisera were tested for reactivity to native sporozoite proteins and for their potential vaccine efficacy using an in vitro inhibition of liver stage development assay in primary human hepatocytes quantified on day 6 post-infection by high content imaging analysis. The anti-PE sera produced significant inhibition of P. vivax sporozoite invasion and liver stage development. This report provides an initial characterization of potential new PE candidates for a future P. vivax vaccine.
Collapse
Affiliation(s)
- Justin Nicholas
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Sai Lata De
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Pongsakorn Thawornpan
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Awtum M. Brashear
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
- Division of Infectious Diseases, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Surendra Kumar Kolli
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Pradeep Annamalai Subramani
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Samantha J. Barnes
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Liwang Cui
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
- Division of Infectious Diseases, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Francis Babila Ntumngia
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - John H. Adams
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| |
Collapse
|
10
|
Martinez FJ, Guillotte-Blisnick M, Huon C, England P, Popovici J, Laude H, Arowas L, Ungeheuer MN, Reimer JM, Carter D, Reed S, Mukherjee P, Chauhan VS, Chitnis CE. Immunogenicity of a Plasmodium vivax vaccine based on the duffy binding protein formulated using adjuvants compatible for use in humans. Sci Rep 2023; 13:13904. [PMID: 37626150 PMCID: PMC10457348 DOI: 10.1038/s41598-023-40043-6] [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: 05/02/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
The invasion of reticulocytes by Plasmodium vivax merozoites is dependent on the interaction of the Plasmodium vivax Duffy Binding Protein (PvDBP) with the Duffy antigen receptor for chemokines (DARC). The N-terminal cysteine-rich region II of PvDBP (PvDBPII), which binds DARC, is a leading P. vivax malaria vaccine candidate. Here, we have evaluated the immunogenicity of recombinant PvDBPII formulated with the adjuvants Matrix-M and GLA-SE in mice. Analysis of the antibody responses revealed comparable ELISA recognition titres as well as similar recognition of native PvDBP in P. vivax schizonts by immunofluorescence assay. Moreover, antibodies elicited by the two adjuvant formulations had similar functional properties such as avidity, isotype profile and inhibition of PvDBPII-DARC binding. Furthermore, the anti-PvDBPII antibodies were able to block the interaction of DARC with the homologous PvDBPII SalI allele as well as the heterologous PvDBPII PvW1 allele from a Thai clinical isolate that is used for controlled human malaria infections (CHMI). The cross-reactivity of these antibodies with PvW1 suggest that immunization with the PvDBPII SalI strain should neutralize reticulocyte invasion by the challenge P. vivax strain PvW1.
Collapse
Affiliation(s)
- Francisco J Martinez
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, 25-28 Rue du Dr. Roux, 75015, Paris, France
| | - 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
| | - Patrick England
- Plate-Forme de Biophysique Moléculaire, CNRS UMR 3528, Institut Pasteur, Université Paris Cité, Paris, France
| | - Jean Popovici
- Malaria Research Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Hélène Laude
- Investigational Clinical Service and Access to Research Bio-Resources (ICAReB), Institut Pasteur, Paris, France
| | - Laurence Arowas
- Investigational Clinical Service and Access to Research Bio-Resources (ICAReB), Institut Pasteur, Paris, France
| | - Marie-Noëlle Ungeheuer
- Investigational Clinical Service and Access to Research Bio-Resources (ICAReB), Institut Pasteur, Paris, France
| | | | - Darrick Carter
- HDT Bio, Seattle, WA, USA
- PAI Life Sciences Inc., Seattle, WA, USA
| | | | | | - Virander S Chauhan
- International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Chetan E Chitnis
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, 25-28 Rue du Dr. Roux, 75015, Paris, France.
| |
Collapse
|
11
|
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: 5.0] [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
|
12
|
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
|
13
|
Owalla TJ, Hergott DEB, Seilie AM, Staubus W, Chavtur C, Chang M, Kublin JG, Egwang TG, Murphy SC. Rethinking detection of pre-existing and intervening Plasmodium infections in malaria clinical trials. Front Immunol 2022; 13:1003452. [PMID: 36203582 PMCID: PMC9531235 DOI: 10.3389/fimmu.2022.1003452] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/22/2022] [Indexed: 02/02/2023] Open
Abstract
Pre-existing and intervening low-density Plasmodium infections complicate the conduct of malaria clinical trials. These infections confound infection detection endpoints, and their immunological effects may detract from intended vaccine-induced immune responses. Historically, these infections were often unrecognized since infrequent and often analytically insensitive parasitological testing was performed before and during trials. Molecular diagnostics now permits their detection, but investigators must weigh the cost, complexity, and personnel demands on the study and the laboratory when scheduling such tests. This paper discusses the effect of pre-existing and intervening, low-density Plasmodium infections on malaria vaccine trial endpoints and the current methods employed for their infection detection. We review detection techniques, that until recently, provided a dearth of cost-effective strategies for detecting low density infections. A recently deployed, field-tested, simple, and cost-effective molecular diagnostic strategy for detecting pre-existing and intervening Plasmodium infections from dried blood spots (DBS) in malaria-endemic settings is discussed to inform new clinical trial designs. Strategies that combine sensitive molecular diagnostic techniques with convenient DBS collections and cost-effective pooling strategies may enable more thorough and informative infection monitoring in upcoming malaria clinical trials and epidemiological studies.
Collapse
Affiliation(s)
- Tonny J. Owalla
- Department of Immunology and Parasitology, Med Biotech Laboratories, Kampala, Uganda
| | - Dianna E. B. Hergott
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, United States
| | - Annette M. Seilie
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Weston Staubus
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Chris Chavtur
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Ming Chang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - James G. Kublin
- Department of Global Health, University of Washington, Seattle, WA, United States,Seattle Malaria Clinical Trials Center, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Thomas G. Egwang
- Department of Immunology and Parasitology, Med Biotech Laboratories, Kampala, Uganda
| | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States,Seattle Malaria Clinical Trials Center, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States,Department of Microbiology, University of Washington, Seattle, WA, United States,*Correspondence: Sean C. Murphy,
| |
Collapse
|
14
|
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
|
15
|
Flannery EL, Kangwanrangsan N, Chuenchob V, Roobsoong W, Fishbaugher M, Zhou K, Billman ZP, Martinson T, Olsen TM, Schäfer C, Campo B, Murphy SC, Mikolajczak SA, Kappe SH, Sattabongkot J. Plasmodium vivax latent liver infection is characterized by persistent hypnozoites, hypnozoite-derived schizonts, and time-dependent efficacy of primaquine. Mol Ther Methods Clin Dev 2022; 26:427-440. [PMID: 36092359 PMCID: PMC9418049 DOI: 10.1016/j.omtm.2022.07.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/31/2022] [Indexed: 01/13/2023]
Abstract
Plasmodium vivax is a malaria-causing pathogen that establishes a dormant form in the liver (the hypnozoite), which can activate weeks, months, or years after the primary infection to cause a relapse, characterized by secondary blood-stage infection. These asymptomatic and undetectable latent liver infections present a significant obstacle to the goal of global malaria eradication. We use a human liver-chimeric mouse model (FRG huHep) to study P. vivax hypnozoite latency and activation in an in vivo model system. Functional activation of hypnozoites and formation of secondary schizonts is demonstrated by first eliminating primary liver schizonts using a schizont-specific antimalarial tool compound, and then measuring recurrence of secondary liver schizonts in the tissue and an increase in parasite RNA within the liver. We also reveal that, while primaquine does not immediately eliminate hypnozoites from the liver, it arrests developing schizonts and prevents activation of hypnozoites, consistent with its clinical activity in humans. Our findings demonstrate that the FRG huHep model can be used to study the biology of P. vivax infection and latency and assess the activity of anti-relapse drugs.
Collapse
Affiliation(s)
- Erika L. Flannery
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98109, USA
- Corresponding author Erika L. Flannery, Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98109, USA.
| | - Niwat Kangwanrangsan
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Vorada Chuenchob
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98109, USA
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Matthew Fishbaugher
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98109, USA
| | - Kevin Zhou
- Department of Laboratory Medicine and Pathology, and Department of Microbiology, University of Washington, Seattle, WA 98115, USA
| | - Zachary P. Billman
- Department of Laboratory Medicine and Pathology, and Department of Microbiology, University of Washington, Seattle, WA 98115, USA
| | - Thomas Martinson
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98109, USA
| | - Tayla M. Olsen
- Department of Laboratory Medicine and Pathology, and Department of Microbiology, University of Washington, Seattle, WA 98115, USA
| | - Carola Schäfer
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98109, USA
| | - Brice Campo
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, and Department of Microbiology, University of Washington, Seattle, WA 98115, USA
| | - Sebastian A. Mikolajczak
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98109, USA
| | - Stefan H.I. Kappe
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98109, USA
- Department of Pediatrics, University of Washington, Seattle, WA 98105, USA
- Corresponding author Stefan H.I. Kappe, Department of Pediatrics, University of Washington, Seattle, WA 98105, USA.
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Corresponding author Jetsumon Sattabongkot, Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
16
|
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
|
17
|
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, Mac Lochlainn DJ, Salkeld J, Guilotte-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. Impact of a blood-stage vaccine on Plasmodium vivax malaria. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.05.27.22275375. [PMID: 35664997 PMCID: PMC9164524 DOI: 10.1101/2022.05.27.22275375] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background There are no licensed vaccines against Plasmodium vivax , the most common cause of malaria outside of Africa. Methods We conducted two Phase I/IIa clinical trials to assess the safety, immunogenicity and efficacy of two vaccines targeting region II of P. vivax Duffy-binding protein (PvDBPII). Recombinant viral vaccines (using ChAd63 and MVA vectors) were administered at 0, 2 months or in a delayed dosing regimen (0, 17, 19 months), whilst a protein/adjuvant formulation (PvDBPII/Matrix-M™) was administered monthly (0, 1, 2 months) or in a delayed dosing regimen (0, 1, 14 months). Delayed regimens were due to trial halts during the COVID-19 pandemic. Volunteers underwent heterologous controlled human malaria infection (CHMI) with blood-stage P. vivax parasites at 2-4 weeks following their last vaccination, alongside unvaccinated controls. Efficacy was assessed by comparison of parasite multiplication rate (PMR) in blood post-CHMI, modelled from parasitemia measured by quantitative polymerase-chain-reaction (qPCR). Results Thirty-two volunteers were enrolled and vaccinated (n=16 for each vaccine). No safety concerns were identified. PvDBPII/Matrix-M™, given in the delayed dosing regimen, elicited the highest antibody responses and reduced the mean PMR following CHMI by 51% (range 36-66%; n=6) compared to unvaccinated controls (n=13). No other vaccine or regimen impacted parasite growth. In vivo growth inhibition of blood-stage P. vivax correlated with functional antibody readouts of vaccine immunogenicity. Conclusions Vaccination of malaria-naïve adults with a delayed booster regimen of PvDBPII/ Matrix-M™ significantly reduces the growth of blood-stage P. vivax . Funded by the European Commission and Wellcome Trust; VAC069, VAC071 and VAC079 ClinicalTrials.gov numbers NCT03797989 , NCT04009096 and NCT04201431 .
Collapse
Affiliation(s)
- Mimi M Hou
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Jordan R Barrett
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, 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
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Amelia M Lias
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Lloyd D W King
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Nick J Edwards
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | | | - Lucy Kingham
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Ian D Poulton
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Baktash Khozoee
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Cyndi Goh
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Dylan J Mac Lochlainn
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Jo Salkeld
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Micheline Guilotte-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 and Tropical Medicine, London, WC1E 7HT, UK
| | | | - Virander S Chauhan
- International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | | | - Sumi Biswas
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Iona J Taylor
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Alison M Lawrie
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Jee-Sun Cho
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Fay L Nugent
- The 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 and 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
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, 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
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Simon J Draper
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| |
Collapse
|
18
|
Aleshnick M, Florez-Cuadros M, Martinson T, Wilder BK. Monoclonal antibodies for malaria prevention. Mol Ther 2022; 30:1810-1821. [PMID: 35395399 PMCID: PMC8979832 DOI: 10.1016/j.ymthe.2022.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/04/2022] [Accepted: 04/01/2022] [Indexed: 11/29/2022] Open
Abstract
Monoclonal antibodies are highly specific proteins that are cloned from a single B cell and bind to a single epitope on a pathogen. These laboratory-made molecules can serve as prophylactics or therapeutics for infectious diseases and have an impressive capacity to modulate the progression of disease, as demonstrated for the first time on a large scale during the COVID-19 pandemic. The high specificity and natural starting point of monoclonal antibodies afford an encouraging safety profile, yet the high cost of production remains a major limitation to their widespread use. While a monoclonal antibody approach to abrogating malaria infection is not yet available, the unique life cycle of the malaria parasite affords many opportunities for such proteins to act, and preliminary research into the efficacy of monoclonal antibodies in preventing malaria infection, disease, and transmission is encouraging. This review examines the current status and future outlook for monoclonal antibodies against malaria in the context of the complex life cycle and varied antigenic targets expressed in the human and mosquito hosts, and provides insight into the strengths and limitations of this approach to curtailing one of humanity’s oldest and deadliest diseases.
Collapse
Affiliation(s)
- Maya Aleshnick
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | | | - Thomas Martinson
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Brandon K Wilder
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA; Department of Parasitology, U.S. Naval Medical Research 6 (NAMRU-6), Lima, Peru
| |
Collapse
|