1
|
Moon RW, Bushell ESC. Not just monkey business. Science 2025; 387:582-583. [PMID: 39913602 DOI: 10.1126/science.adv2328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
Functional genomics in malaria unlocks comparative biology across the family tree.
Collapse
Affiliation(s)
- Robert W Moon
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Ellen S C Bushell
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
| |
Collapse
|
2
|
Kochayoo P, Moriyama S, Kotaki R, Thawornpan P, Malee C, Leepiyasakulchai C, Ntumngia FB, Adams JH, Takahashi Y, Chootong P. Atypical memory B cells from natural malaria infection produced broadly neutralizing antibodies against Plasmodium vivax variants. PLoS Pathog 2025; 21:e1012866. [PMID: 39847574 PMCID: PMC11756785 DOI: 10.1371/journal.ppat.1012866] [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: 07/31/2024] [Accepted: 12/30/2024] [Indexed: 01/25/2025] Open
Abstract
Expansion of atypical memory B cells (aMBCs) was demonstrated in malaria-exposed individuals. To date, the generation of P. vivax-specific aMBCs and their function in protective humoral immune responses is unknown. Here, P. vivax Duffy Binding Protein II (PvDBPII) probes were generated to detect the development and durability of specific aMBCs, and to demonstrate the capacity of these cells to produce neutralizing antibodies following natural infections. PvDBPII-specific aMBCs were elicited during malaria illness, and they persisted through the recovery phase of infections. To address biology and function of P. vivax-specific aMBCs in producing protective antibodies, a single MBC was cultured, and the secreted IgG was tested for binding and inhibition activity. The aMBC-derived clones produced antibodies with variable levels of anti-PvDBPII IgG in cultures, and some produced high antibody levels comparable to classical MBC clones. Thus, we focused our attention on the function of aMBCs in producing neutralizing antibodies. Among the aMBC clones, A1F12 and B4E11 produced broadly neutralizing antibodies against a panel of PvDBPII variants. Notably, B cell receptors (BCRs) of PvDBPII-specific aMBCs expressed unique IGHV genes, with similar usage of IGHV1-3, comparable to classical MBCs. The somatic hypermutation (SHM) rate and CDR3 length of VH and Vκ in these two MBC subsets were not significantly different. Together, our findings revealed that P. vivax infections elicited the development and persistence of P. vivax-specific aMBCs. The accumulation of aMBCs during and following infections might play an important role in producing protective antibodies against malaria.
Collapse
Affiliation(s)
- Piyawan Kochayoo
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Saya Moriyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
| | - Ryutaro Kotaki
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
| | - Pongsakorn Thawornpan
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Chayapat Malee
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Chaniya Leepiyasakulchai
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Francis Babila Ntumngia
- Center for Global Health and Inter-Disciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - John H. Adams
- Center for Global Health and Inter-Disciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
- Institute for Vaccine Research and Development, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| |
Collapse
|
3
|
Yanik S, Yu H, Chaiyawong N, Adewale-Fasoro O, Dinis LR, Narayanasamy RK, Lee EC, Lubonja A, Li B, Jaeger S, Srinivasan P. Application of Machine Learning in a Rodent Malaria Model for Rapid, Accurate, and Consistent Parasite Counts. Am J Trop Med Hyg 2024; 111:967-976. [PMID: 39255803 PMCID: PMC11542515 DOI: 10.4269/ajtmh.24-0135] [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: 02/27/2024] [Accepted: 06/04/2024] [Indexed: 09/12/2024] Open
Abstract
Rodent malaria models serve as important preclinical antimalarial and vaccine testing tools. Evaluating treatment outcomes in these models often requires manually counting parasite-infected red blood cells (iRBCs), a time-consuming process, which can be inconsistent between individuals and laboratories. We have developed an easy-to-use machine learning (ML)-based software, Malaria Screener R, to expedite and standardize such studies by automating the counting of Plasmodium iRBCs in rodents. This software can process Giemsa-stained blood smear images captured by any camera-equipped microscope. It features an intuitive graphical user interface that facilitates image processing and visualization of the results. The software has been developed as a desktop application that processes images on standard Windows and MacOS computers. A previous ML model created by the authors designed to count Plasmodium falciparum-infected human RBCs did not perform well counting Plasmodium-infected mouse RBCs. We leveraged that model by loading the pretrained weights and training the algorithm with newly collected data to target Plasmodium yoelii- and Plasmodium berghei-infected mouse RBCs. This new model reliably measured both P. yoelii and P. berghei parasitemia (R2 = 0.9916). Additional rounds of training data to incorporate variances due to length of Giemsa staining and type of microscopes, etc., have produced a generalizable model, meeting WHO competency level 1 for the subcategory of parasite counting using independent microscopes. Reliable, automated analyses of blood-stage parasitemia will facilitate rapid and consistent evaluation of novel vaccines and antimalarials across laboratories in an easily accessible in vivo malaria model.
Collapse
Affiliation(s)
- Sean Yanik
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, Maryland
- Malaria Research Institute, Johns Hopkins School of Public Health, Baltimore, Maryland
| | - Hang Yu
- National Library of Medicine, National Institutes of Health, Bethesda, Maryland
| | - Nattawat Chaiyawong
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, Maryland
- Malaria Research Institute, Johns Hopkins School of Public Health, Baltimore, Maryland
| | - Opeoluwa Adewale-Fasoro
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, Maryland
- Malaria Research Institute, Johns Hopkins School of Public Health, Baltimore, Maryland
| | - Luciana Ribeiro Dinis
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, Maryland
- Malaria Research Institute, Johns Hopkins School of Public Health, Baltimore, Maryland
| | - Ravi Kumar Narayanasamy
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, Maryland
- Malaria Research Institute, Johns Hopkins School of Public Health, Baltimore, Maryland
| | - Elizabeth C. Lee
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, Maryland
- Malaria Research Institute, Johns Hopkins School of Public Health, Baltimore, Maryland
| | - Ariel Lubonja
- Department of Computer Science, Johns Hopkins Whiting School of Engineering, Baltimore, Maryland
| | - Bowen Li
- Department of Computer Science, Johns Hopkins Whiting School of Engineering, Baltimore, Maryland
| | - Stefan Jaeger
- National Library of Medicine, National Institutes of Health, Bethesda, Maryland
| | - Prakash Srinivasan
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, Maryland
- Malaria Research Institute, Johns Hopkins School of Public Health, Baltimore, Maryland
| |
Collapse
|
4
|
Wall RJ, MacGowan SA, Hallyburton I, Syed AJ, Ajay Castro S, Dey G, Milne R, Patterson S, Phelan J, Wiedemar N, Wyllie S. ResMAP-a saturation mutagenesis platform enabling parallel profiling of target-specific resistance-conferring mutations in Plasmodium. mBio 2024; 15:e0170824. [PMID: 39191404 PMCID: PMC11481570 DOI: 10.1128/mbio.01708-24] [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: 06/04/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
New and improved drugs are required for the treatment and ultimate eradication of malaria. The efficacy of front-line therapies is now threatened by emerging drug resistance; thus, new tools to support the development of drugs with a lower propensity for resistance are needed. Here, we describe the development of a RESistance Mapping And Profiling (ResMAP) platform for the identification of resistance-conferring mutations in Plasmodium drug targets. Proof-of-concept studies focused on interrogating the antimalarial drug target, Plasmodium falciparum lysyl tRNA synthetase (PfKRS). Saturation mutagenesis was used to construct a plasmid library encoding all conceivable mutations within a 20-residue span at the base of the PfKRS ATP-binding pocket. The superior transfection efficiency of Plasmodium knowlesi was exploited to generate a high coverage parasite library expressing PfKRS bearing all possible amino acid changes within this region of the enzyme. The selection of the library with PfKRS inhibitors, cladosporin and DDD01510706, successfully identified multiple resistance-conferring substitutions. Genetic validation of a subset of these mutations confirmed their direct role in resistance, with computational modeling used to dissect the structural basis of resistance. The application of ResMAP to inform the development of resistance-resilient antimalarials of the future is discussed. IMPORTANCE An increase in treatment failures for malaria highlights an urgent need for new tools to understand and minimize the spread of drug resistance. We describe the development of a RESistance Mapping And Profiling (ResMAP) platform for the identification of resistance-conferring mutations in Plasmodium spp, the causative agent of malaria. Saturation mutagenesis was used to generate a mutation library containing all conceivable mutations for a region of the antimalarial-binding site of a promising drug target, Plasmodium falciparum lysyl tRNA synthetase (PfKRS). Screening of this high-coverage library with characterized PfKRS inhibitors revealed multiple resistance-conferring substitutions including several clinically relevant mutations. Genetic validation of these mutations confirmed resistance of up to 100-fold and computational modeling dissected their role in drug resistance. We discuss potential applications of this data including the potential to design compounds that can bypass the most serious resistance mutations and future resistance surveillance.
Collapse
Affiliation(s)
- Richard J. Wall
- Wellcome Center for Anti-infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dow Street, Dundee, United Kingdom
| | - Stuart A. MacGowan
- Division of Computational Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Irene Hallyburton
- Drug Discovery Unit, Wellcome Center for Anti-infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, United Kingdom
| | - Aisha J. Syed
- Wellcome Center for Anti-infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dow Street, Dundee, United Kingdom
| | - Sowmya Ajay Castro
- Division of Molecular Microbiology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Gourav Dey
- Wellcome Center for Anti-infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dow Street, Dundee, United Kingdom
| | - Rachel Milne
- Wellcome Center for Anti-infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dow Street, Dundee, United Kingdom
| | - Stephen Patterson
- Wellcome Center for Anti-infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dow Street, Dundee, United Kingdom
| | - Jody Phelan
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Natalie Wiedemar
- Wellcome Center for Anti-infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dow Street, Dundee, United Kingdom
| | - Susan Wyllie
- Wellcome Center for Anti-infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dow Street, Dundee, United Kingdom
| |
Collapse
|
5
|
Barrett JR, Pipini D, Wright ND, Cooper AJR, Gorini G, Quinkert D, Lias AM, Davies H, Rigby CA, Aleshnick M, Williams BG, Bradshaw WJ, Paterson NG, Martinson T, Kirtley P, Picard L, Wiggins CD, Donnellan FR, King LDW, Wang LT, Popplewell JF, Silk SE, de Ruiter Swain J, Skinner K, Kotraiah V, Noe AR, MacGill RS, King CR, Birkett AJ, Soisson LA, Minassian AM, Lauffenburger DA, Miura K, Long CA, Wilder BK, Koekemoer L, Tan J, Nielsen CM, McHugh K, Draper SJ. Analysis of the diverse antigenic landscape of the malaria protein RH5 identifies a potent vaccine-induced human public antibody clonotype. Cell 2024; 187:4964-4980.e21. [PMID: 39059380 PMCID: PMC11380582 DOI: 10.1016/j.cell.2024.06.015] [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: 09/28/2023] [Revised: 04/14/2024] [Accepted: 06/10/2024] [Indexed: 07/28/2024]
Abstract
The highly conserved and essential Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5) has emerged as the leading target for vaccines against the disease-causing blood stage of malaria. However, the features of the human vaccine-induced antibody response that confer highly potent inhibition of malaria parasite invasion into red blood cells are not well defined. Here, we characterize 236 human IgG monoclonal antibodies, derived from 15 donors, induced by the most advanced PfRH5 vaccine. We define the antigenic landscape of this molecule and establish that epitope specificity, antibody association rate, and intra-PfRH5 antibody interactions are key determinants of functional anti-parasitic potency. In addition, we identify a germline IgG gene combination that results in an exceptionally potent class of antibody and demonstrate its prophylactic potential to protect against P. falciparum parasite challenge in vivo. This comprehensive dataset provides a framework to guide rational design of next-generation vaccines and prophylactic antibodies to protect against blood-stage malaria.
Collapse
Affiliation(s)
- Jordan R Barrett
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Dimitra Pipini
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Nathan D Wright
- Centre for Medicines Discovery, University of Oxford, Oxford OX3 7FZ, UK
| | - Andrew J R Cooper
- Antibody Biology Unit, Laboratory of Immunogenetics, NIAID/NIH, Rockville, MD 20852, USA
| | - Giacomo Gorini
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Doris Quinkert
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, 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; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Hannah Davies
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Cassandra A Rigby
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Maya Aleshnick
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Barnabas G Williams
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - William J Bradshaw
- Centre for Medicines Discovery, University of Oxford, Oxford OX3 7FZ, UK
| | - Neil G Paterson
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, Oxfordshire OX11 0DE, UK
| | - Thomas Martinson
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Payton Kirtley
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Luc Picard
- Department of Biological Engineering, MIT, Cambridge, MA, USA
| | | | - Francesca R Donnellan
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, 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; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Lawrence T Wang
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; Antibody Biology Unit, Laboratory of Immunogenetics, NIAID/NIH, Rockville, MD 20852, USA
| | | | - Sarah E Silk
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Jed de Ruiter Swain
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Katherine Skinner
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Amy R Noe
- Leidos Life Sciences, Frederick, MD, USA
| | - Randall S MacGill
- Center for Vaccine Innovation and Access, PATH, Washington, DC 20001, USA
| | - C Richter King
- Center for Vaccine Innovation and Access, PATH, Washington, DC 20001, USA
| | - Ashley J Birkett
- Center for Vaccine Innovation and Access, PATH, Washington, DC 20001, USA
| | | | - Angela M Minassian
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | | | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Brandon K Wilder
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Lizbé Koekemoer
- Centre for Medicines Discovery, University of Oxford, Oxford OX3 7FZ, UK
| | - Joshua Tan
- Antibody Biology Unit, Laboratory of Immunogenetics, NIAID/NIH, Rockville, MD 20852, USA
| | - Carolyn M Nielsen
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Kirsty McHugh
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, 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; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
6
|
Dickey TH, McAleese H, Salinas ND, Lambert LE, Tolia NH. Structure-based design of a Plasmodium vivax Duffy-binding protein immunogen focuses the antibody response to functional epitopes. Protein Sci 2024; 33:e5095. [PMID: 38988315 PMCID: PMC11237555 DOI: 10.1002/pro.5095] [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: 02/07/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 07/12/2024]
Abstract
The Duffy-binding protein (DBP) is a promising antigen for a malaria vaccine that would protect against clinical symptoms caused by Plasmodium vivax infection. Region II of DBP (DBP-II) contains the receptor-binding domain that engages host red blood cells, but DBP-II vaccines elicit many non-neutralizing antibodies that bind distal to the receptor-binding surface. Here, we engineered a truncated DBP-II immunogen that focuses the immune response to the receptor-binding surface. This immunogen contains the receptor-binding subdomain S1S2 and lacks the immunodominant subdomain S3. Structure-based computational design of S1S2 identified combinatorial amino acid changes that stabilized the isolated S1S2 without perturbing neutralizing epitopes. This immunogen elicited DBP-II-specific antibodies in immunized mice that were significantly enriched for blocking activity compared to the native DBP-II antigen. This generalizable design process successfully stabilized an integral core fragment of a protein and focused the immune response to desired epitopes to create a promising new antigen for malaria vaccine development.
Collapse
MESH Headings
- Protozoan Proteins/immunology
- Protozoan Proteins/chemistry
- Protozoan Proteins/genetics
- Antigens, Protozoan/immunology
- Antigens, Protozoan/chemistry
- Antigens, Protozoan/genetics
- Plasmodium vivax/immunology
- Animals
- Malaria Vaccines/immunology
- Malaria Vaccines/chemistry
- Epitopes/immunology
- Epitopes/chemistry
- Mice
- Antibodies, Protozoan/immunology
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/genetics
- Models, Molecular
- Malaria, Vivax/immunology
- Malaria, Vivax/prevention & control
- Mice, Inbred BALB C
Collapse
Affiliation(s)
- Thayne H. Dickey
- Host‐Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health (NIH)BethesdaMarylandUSA
| | - Holly McAleese
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health (NIH)BethesdaMarylandUSA
| | - Nichole D. Salinas
- Host‐Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health (NIH)BethesdaMarylandUSA
| | - Lynn E. Lambert
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health (NIH)BethesdaMarylandUSA
| | - Niraj H. Tolia
- Host‐Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health (NIH)BethesdaMarylandUSA
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Yanik S, Yu H, Chaiyawong N, Adewale-Fasoro O, Dinis LR, Narayanasamy RK, Lee EC, Lubonja A, Li B, Jaeger S, Srinivasan P. Application of machine learning in a rodent malaria model for rapid, accurate, and consistent parasite counts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597554. [PMID: 38895284 PMCID: PMC11185661 DOI: 10.1101/2024.06.05.597554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Rodent malaria models serve as important preclinical antimalarial and vaccine testing tools. Evaluating treatment outcomes in these models often requires manually counting parasite-infected red blood cells (iRBCs), a time-consuming process, which can be inconsistent between individuals and labs. We have developed an easy-to-use machine learning (ML)-based software, Malaria Screener R, to expedite and standardize such studies by automating the counting of Plasmodium iRBCs in rodents. This software can process Giemsa-stained blood smear images captured by any camera-equipped microscope. It features an intuitive graphical user interface that facilitates image processing and visualization of the results. The software has been developed as a desktop application that processes images on standard Windows and Mac OS computers. A previous ML model created by the authors designed to count P. falciparum -infected human RBCs did not perform well counting Plasmodium -infected mouse RBCs. We leveraged that model by loading the pre-trained weights and training the algorithm with newly collected data to target P. yoelii and P. berghei mouse iRBCs. This new model reliably measured both P. yoelii and P. berghei parasitemia (R 2 = 0.9916). Additional rounds of training data to incorporate variances due to length of Giemsa staining, microscopes etc, have produced a generalizable model, meeting WHO Competency Level 1 for the sub-category of parasite counting using independent microscopes. Reliable, automated analyses of blood-stage parasitemia will facilitate rapid and consistent evaluation of novel vaccines and antimalarials across labs in an easily accessible in vivo malaria model.
Collapse
|
10
|
Little E, Shenkutie TT, Negash MT, Abagero BR, Abebe A, Popovici J, Feleke SM, Lo E. Prevalence and Characteristics of Plasmodium vivax Gametocytes in Duffy-Positive and Duffy-Negative Populations across Ethiopia. Am J Trop Med Hyg 2024; 110:1091-1099. [PMID: 38626749 PMCID: PMC11154031 DOI: 10.4269/ajtmh.23-0877] [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: 12/09/2023] [Accepted: 01/04/2024] [Indexed: 04/18/2024] Open
Abstract
Plasmodium parasites replicate asexually in human hosts. The proportion of infections that carries gametocytes is a proxy for human-to-mosquito transmissibility. It is unclear which proportion of Plasmodium vivax infections in Duffy-negative populations carries gametocytes. We determined the prevalence and characteristics of P. vivax gametocytes in Duffy-positive and -negative populations across broad regions of Ethiopia. Finger-prick blood samples were collected for microscopic and molecular screening of Plasmodium parasites and Duffy status of individuals. Molecular screening of Plasmodium species and Duffy blood group genotyping was done using SYBR green and the Taqman quantitative polymerase chain reaction method. Of the 447 febrile patients who were shown to be P. vivax smear positive, 414 (92.6%) were confirmed by molecular screening as P. vivax and 16 (3.9%) of them were from Duffy-negative individuals. Of these, 5 of 16 (31.3%) Duffy-negative P. vivax-infected samples were detected with gametocytes. Of the 398 Duffy-positive P. vivax-infected samples, 150 (37.7%) were detected with gametocytes, slightly greater than that in Duffy-negative samples. This study highlights the presence of P. vivax gametocytes in Duffy-negative infections, suggestive of human-to-mosquito transmissibility. Although P. vivax infections in Duffy-negative individuals were commonly associated with low parasitemia, some of these infections were shown to have relatively high parasitemia and may represent a prominent erythrocyte invasion capability of P. vivax, and hidden reservoirs that can contribute to transmission. A better understanding of P. vivax transmission biology and gametocyte function particularly in Duffy-negative populations would aid future treatment and management of P. vivax malaria in Africa.
Collapse
Affiliation(s)
- Ebony Little
- Department of Biological Sciences, University of North Carolina at Charlotte, North Carolina
| | - Tassew T. Shenkutie
- Department of Microbiology and Immunology, Drexel University, College of Medicine, Philadelphia, Pennsylvania
- Department of Medical Laboratory Sciences, Debre Brehan University, Ethiopia
| | | | - Beka R. Abagero
- Department of Molecular and Cellular Biology and Genetics, Drexel University, College of Medicine, Philadelphia, Pennsylvania
| | - Abnet Abebe
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | | | | | - Eugenia Lo
- Department of Biological Sciences, University of North Carolina at Charlotte, North Carolina
- Department of Microbiology and Immunology, Drexel University, College of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
11
|
Ma R, Salinas ND, Orr-Gonzalez S, Richardson B, Ouahes T, Torano H, Jenkins BJ, Dickey TH, Neal J, Duan J, Morrison RD, Gittis AG, Doritchamou JYA, Zaidi I, Lambert LE, Duffy PE, Tolia NH. Structure-guided design of VAR2CSA-based immunogens and a cocktail strategy for a placental malaria vaccine. PLoS Pathog 2024; 20:e1011879. [PMID: 38437239 PMCID: PMC10939253 DOI: 10.1371/journal.ppat.1011879] [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: 08/14/2023] [Revised: 03/14/2024] [Accepted: 11/29/2023] [Indexed: 03/06/2024] Open
Abstract
Placental accumulation of Plasmodium falciparum infected erythrocytes results in maternal anemia, low birth weight, and pregnancy loss. The parasite protein VAR2CSA facilitates the accumulation of infected erythrocytes in the placenta through interaction with the host receptor chondroitin sulfate A (CSA). Antibodies that prevent the VAR2CSA-CSA interaction correlate with protection from placental malaria, and VAR2CSA is a high-priority placental malaria vaccine antigen. Here, structure-guided design leveraging the full-length structures of VAR2CSA produced a stable immunogen that retains the critical conserved functional elements of VAR2CSA. The design expressed with a six-fold greater yield than the full-length protein and elicited antibodies that prevent adhesion of infected erythrocytes to CSA. The reduced size and adaptability of the designed immunogen enable efficient production of multiple variants of VAR2CSA for use in a cocktail vaccination strategy to increase the breadth of protection. These designs form strong foundations for the development of potent broadly protective placental malaria vaccines.
Collapse
Affiliation(s)
- Rui Ma
- 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, Maryland, United States of America
| | - 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, Maryland, United States of America
| | - Sachy Orr-Gonzalez
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Brandi Richardson
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Tarik Ouahes
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Holly Torano
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Bethany J Jenkins
- Pathogenesis and Immunity Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - 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, Maryland, United States of America
| | - Jillian Neal
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Junhui Duan
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Robert D Morrison
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Apostolos G Gittis
- Structural Biology Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Justin Y A Doritchamou
- Pathogenesis and Immunity Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Irfan Zaidi
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lynn E Lambert
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Patrick E Duffy
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- Pathogenesis and Immunity Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - 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, Maryland, United States of America
| |
Collapse
|
12
|
Little E, Shenkutie TT, Negash MT, Abagero BR, Abebe A, Popovici J, Mekasha S, Lo E. Prevalence and characteristics of Plasmodium vivax Gametocytes in Duffy-positive and Duffy-negative populations across Ethiopia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.10.23299780. [PMID: 38168152 PMCID: PMC10760292 DOI: 10.1101/2023.12.10.23299780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Plasmodium parasites replicate asexually in the human host. The proportion of infections that carries gametocytes is a proxy for human-to-mosquito transmissibility. It is unclear what proportion of P. vivax infections in Duffy-negatives carries gametocytes. This study aims to determine the prevalence of P. vivax in Duffy-negatives across broad regions of Ethiopia and characterize parasite stages. Finger-prick blood samples were collected for microscopic and molecular screening of Plasmodium parasites and Duffy status of individuals. Molecular screening of plasmodium species and Duffy blood group genotyping was done using SYBR green and Taqman qPCR method. Among the total 447 samples, 414 (92.6%) were P. vivax confirmed and, 16 (3.9%) of them were from Duffy-negatives. Of these, 5/16 (31.3%) Duffy-negative P. vivax-infected samples were detected with gametocytes. Of the 398 Duffy-positive P. vivax-infected samples, 150 (37.7%) were detected with gametocytes, slightly higher than that in Duffy-negatives. This study highlights the presence of P. vivax gametocytes in Duffy-negative infections, suggestive of human-to-mosquito transmissibility. Although P. vivax infections in Duffy-negatives are commonly associated with low parasitemia, some of these infections were shown with relatively high parasitemia and may represent better erythrocyte invasion capability of P. vivax and hidden reservoirs that can contribute to transmission. A better understanding of P. vivax transmission biology and gametocyte function particularly in Duffy-negative populations would aid future treatment and management of vivax malaria in Africa.
Collapse
Affiliation(s)
- Ebony Little
- Department of Biological Sciences, University of North Carolina at Charlotte, North Carolina, USA
| | - Tassew T. Shenkutie
- Department of Microbiology and Immunology, Drexel University, College of Medicine, Philadelphia, PA, USA
- Department of Medical Laboratory Sciences, Debre Brehan University, Debre Brehan, Ethiopia
| | | | - Beka R. Abagero
- Department of Molecular and Cellular Biology and Genetics, Drexel University, College of Medicine, Philadelphia, PA, USA
| | - Abnet Abebe
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | | | - Sindew Mekasha
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Eugenia Lo
- Department of Biological Sciences, University of North Carolina at Charlotte, North Carolina, USA
- Department of Microbiology and Immunology, Drexel University, College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
13
|
Watson QD, Carias LL, Malachin A, Redinger KR, Bosch J, Bardelli M, Baldor L, Feufack-Donfack LB, Popovici J, Moon RW, Draper SJ, Zimmerman PA, King CL. Human monoclonal antibodies inhibit invasion of transgenic Plasmodium knowlesi expressing Plasmodium vivax Duffy binding protein. Malar J 2023; 22:369. [PMID: 38049801 PMCID: PMC10696754 DOI: 10.1186/s12936-023-04766-1] [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: 07/10/2023] [Accepted: 10/24/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Plasmodium vivax has been more resistant to various control measures than Plasmodium falciparum malaria because of its greater transmissibility and ability to produce latent parasite forms. Therefore, developing P. vivax vaccines and therapeutic monoclonal antibodies (humAbs) remains a high priority. The Duffy antigen receptor for chemokines (DARC) expressed on erythrocytes is central to P. vivax invasion of reticulocytes. P. vivax expresses a Duffy binding protein (PvDBP) on merozoites, a DARC ligand, and the DARC: PvDBP interaction is critical for P. vivax blood stage malaria. Therefore, PvDBP is a leading vaccine candidate for P. vivax and a target for therapeutic human monoclonal antibodies (humAbs). METHODS Here, the functional activity of humAbs derived from naturally exposed and vaccinated individuals are compared for the first time using easily cultured Plasmodium knowlesi (P. knowlesi) that had been genetically modified to replace its endogenous PkDBP orthologue with PvDBP to create a transgenic parasite, PkPvDBPOR. This transgenic parasite requires DARC to invade human erythrocytes but is not reticulocyte restricted. This model was used to evaluate the invasion inhibition potential of 12 humAbs (9 naturally acquired; 3 vaccine-induced) targeting PvDBP individually and in combinations using growth inhibition assays (GIAs). RESULTS The PvDBP-specific humAbs demonstrated 70-100% inhibition of PkPvDBPOR invasion with the IC50 values ranging from 51 to 338 µg/mL for the 9 naturally acquired (NA) humAbs and 33 to 99 µg/ml for the 3 vaccine-induced (VI) humAbs. To evaluate antagonistic, additive, or synergistic effects, six pairwise combinations were performed using select humAbs. Of these combinations tested, one NA/NA (099100/094083) combination demonstrated relatively strong additive inhibition between 10 and 100 µg/mL; all combinations of NA and VI humAbs showed additive inhibition at concentrations below 25 µg/mL and antagonism at higher concentrations. None of the humAb combinations showed synergy. Invasion inhibition efficacy by some mAbs shown with PkPvDBPOR was closely replicated using P. vivax clinical isolates. CONCLUSION The PkPvDBPOR transgenic model is a robust surrogate of P. vivax to assess invasion and growth inhibition of human monoclonal Abs recognizing PvDBP individually and in combination. There was no synergistic interaction for growth inhibition with the humAbs tested here that target different epitopes or subdomains of PvDBP, suggesting little benefit in clinical trials using combinations of these humAbs.
Collapse
Affiliation(s)
- Quentin D Watson
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Lenore L Carias
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Alyssa Malachin
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Karli R Redinger
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jürgen Bosch
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | - Lea Baldor
- Malaria Research Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | | | - Jean Popovici
- Malaria Research Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Robert W Moon
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Simon J Draper
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Peter A Zimmerman
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| | - Christopher L King
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Veterans Affairs Medical Center, Cleveland, OH, USA.
| |
Collapse
|
14
|
Dickey TH, Tolia NH. Designing an effective malaria vaccine targeting Plasmodium vivax Duffy-binding protein. Trends Parasitol 2023; 39:850-858. [PMID: 37481347 PMCID: PMC11099547 DOI: 10.1016/j.pt.2023.06.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/24/2023]
Abstract
Malaria caused by the Plasmodium vivax parasite is a major global health burden. Immunity against blood-stage infection reduces parasitemia and disease severity. Duffy-binding protein (DBP) is the primary parasite protein responsible for the invasion of red blood cells and it is a leading subunit vaccine candidate. An effective vaccine, however, is still lacking despite decades of interest in DBP as a vaccine candidate. This review discusses the reasons for targeting DBP, the challenges associated with developing a vaccine, and modern structural vaccinology methods that could be used to create an effective DBP vaccine. Next-generation DBP vaccines have the potential to elicit a broadly protective immune response and provide durable and potent protection from P. vivax malaria.
Collapse
Affiliation(s)
- Thayne H Dickey
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20894, USA
| | - Niraj H Tolia
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20894, USA.
| |
Collapse
|
15
|
Dash R, Skillman KM, Pereira L, Mascarenhas A, Dass S, Walke J, Almeida A, Fernandes M, Gomes E, White J, Chery-Karschney L, Khandeparkar A, Rathod PK, Duraisingh MT, Kanjee U. Development of a Plasmodium vivax biobank for functional ex vivo assays. Malar J 2023; 22:250. [PMID: 37653486 PMCID: PMC10470152 DOI: 10.1186/s12936-023-04668-2] [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: 03/17/2023] [Accepted: 08/07/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND Plasmodium vivax is the second most prevalent cause of malaria yet remains challenging to study due to the lack of a continuous in vitro culture system, highlighting the need to establish a biobank of clinical isolates with multiple freezes per sample for use in functional assays. Different methods for cryopreserving parasite isolates were compared and subsequently the most promising one was validated. Enrichment of early- and late-stage parasites and parasite maturation were quantified to facilitate assay planning. METHODS In order to compare cryopreservation protocols, nine clinical P. vivax isolates were frozen with four glycerolyte-based mixtures. Parasite recovery post thaw, post KCl-Percoll enrichment and in short-term in vitro culture was measured via slide microscopy. Enrichment of late-stage parasites by magnetic activated cell sorting (MACS) was measured. Short and long-term storage of parasites at either - 80 °C or liquid nitrogen were also compared. RESULTS Of the four cryopreservation mixtures, one mixture (glycerolyte:serum:RBC at a 2.5:1.5:1 ratio) resulted in improved parasite recovery and statistically significant (P < 0.05) enhancement in parasite survival in short-term in vitro culture. A parasite biobank was subsequently generated using this protocol resulting in a collection of 106 clinical isolates, each with 8 vials. The quality of the biobank was validated by measuring several factors from 47 thaws: the average reduction in parasitaemia post-thaw (25.3%); the average fold enrichment post KCl-Percoll (6.65-fold); and the average percent recovery of parasites (22.0%, measured from 30 isolates). During short-term in vitro culture, robust maturation of ring stage parasites to later stages (> 20% trophozoites, schizonts and gametocytes) was observed in 60.0% of isolates by 48 h. Enrichment of mature parasite stages via MACS showed good reproducibility, with an average of 30.0% post-MACS parasitaemia and an average of 5.30 × 105 parasites/vial. Finally, the effect of storage temperature was tested, and no large impacts from short-term (7 days) or long-term (7-10 years) storage at - 80 °C on parasite recovery, enrichment or viability was observed. CONCLUSIONS Here, an optimized freezing method for P. vivax clinical isolates is demonstrated as a template for the generation and validation of a parasite biobank for use in functional assays.
Collapse
Affiliation(s)
- Rashmi Dash
- Goa Medical College and Hospital, Bambolim, Goa, 403202, India
- Departments of Chemistry and Global Health, University of Washington, Seattle, WA, 98195, USA
| | - Kristen M Skillman
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Ligia Pereira
- Goa Medical College and Hospital, Bambolim, Goa, 403202, India
- Departments of Chemistry and Global Health, University of Washington, Seattle, WA, 98195, USA
| | - Anjali Mascarenhas
- Goa Medical College and Hospital, Bambolim, Goa, 403202, India
- Departments of Chemistry and Global Health, University of Washington, Seattle, WA, 98195, USA
| | - Sheena Dass
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Jayashri Walke
- Goa Medical College and Hospital, Bambolim, Goa, 403202, India
- Departments of Chemistry and Global Health, University of Washington, Seattle, WA, 98195, USA
| | - Anvily Almeida
- Goa Medical College and Hospital, Bambolim, Goa, 403202, India
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Mezia Fernandes
- Goa Medical College and Hospital, Bambolim, Goa, 403202, India
- Departments of Chemistry and Global Health, University of Washington, Seattle, WA, 98195, USA
| | - Edwin Gomes
- Goa Medical College and Hospital, Bambolim, Goa, 403202, India
| | - John White
- Departments of Chemistry and Global Health, University of Washington, Seattle, WA, 98195, USA
| | - Laura Chery-Karschney
- Departments of Chemistry and Global Health, University of Washington, Seattle, WA, 98195, USA
| | | | - Pradipsinh K Rathod
- Departments of Chemistry and Global Health, University of Washington, Seattle, WA, 98195, USA
| | - Manoj T Duraisingh
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Usheer Kanjee
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA.
| |
Collapse
|
16
|
Ren Z, Shi Q, Xu S, Xu J, Yin Y, Lin Z, Xu S, Ma X, Liu Y, Zhu G, He X, Lu J, Li Y, Zhang W, Liu J, Yang Y, Han ET, Cao J, Lu F. Elicitation of T-cell-derived IFN-γ-dependent immunity by highly conserved Plasmodium ovale curtisi Duffy binding protein domain region II (PocDBP-RII). Parasit Vectors 2023; 16:269. [PMID: 37553591 PMCID: PMC10410920 DOI: 10.1186/s13071-023-05897-9] [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: 05/24/2023] [Accepted: 07/27/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Infections with Plasmodium ovale are widely distributed but rarely investigated, and the resulting burden of disease has been underestimated. Plasmodium ovale curtisi Duffy binding protein domain region II (PocDBP-RII) is an essential ligand for reticulocyte recognition and host cell invasion by P. ovale curtisi. However, the genomic variation, antigenicity and immunogenicity of PocDBP-RII remain major knowledge gaps. METHODS A total of 93 P. ovale curtisi samples were collected from migrant workers who returned to China from 17 countries in Africa between 2012 and 2016. The genetic polymorphism, natural selection and copy number variation (CNV) were investigated by sequencing and real-time PCR. The antigenicity and immunogenicity of the recombinant PocDBP-RII (rPocDBP-RII) protein were further examined, and the humoral and cellular responses of immunized mice were assessed using protein microarrays and flow cytometry. RESULTS Efficiently expressed and purified rPocDBP-RII (39 kDa) was successfully used as an antigen for immunization in mice. The haplotype diversity (Hd) of PocDBP-RII gene was 0.105, and the nucleotide diversity index (π) was 0.00011. No increased copy number was found among the collected isolates of P. ovale curtisi. Furthermore, rPocDBP-RII induced persistent antigen-specific antibody production with a serum IgG antibody titer of 1: 16,000. IFN-γ-producing T cells, rather than IL-10-producing cells, were activated in response to the stimulation of rPocDBP-RII. Compared to PBS-immunized mice (negative control), there was a higher percentage of CD4+CD44highCD62L- T cells (effector memory T cells) and CD8+CD44highCD62L+ T cells (central memory T cells) in rPocDBP-RII‑immunized mice. CONCLUSIONS PocDBP-RII sequences were highly conserved in clinical isolates of P. ovale curtisi. rPocDBP-RII protein could mediate protective blood-stage immunity through IFN-γ-producing CD4+ and CD8+ T cells and memory T cells, in addition to inducing specific antibodies. Our results suggested that rPocDBP-RII protein has potential as a vaccine candidate to provide assessment and guidance for malaria control and elimination activities.
Collapse
Affiliation(s)
- Zhenyu Ren
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Qiyang Shi
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Simin Xu
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
- Changshu Second People's Hospital, Suzhou, 215500, Jiangsu, People's Republic of China
| | - Jiahui Xu
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Yi Yin
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Zhijie Lin
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Sui Xu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Xiaoqin Ma
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Yaobao Liu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Guoding Zhu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Xinlong He
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Jingyuan Lu
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Yinyue Li
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Wenwen Zhang
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Jiali Liu
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Yun Yang
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Jun Cao
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China.
| | - Feng Lu
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.
- Affiliated Hospital of Yangzhou University, Yangzhou, 225000, People's Republic of China.
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.
| |
Collapse
|
17
|
Hart MN, Mohring F, DonVito SM, Thomas JA, Muller-Sienerth N, Wright GJ, Knuepfer E, Saibil HR, Moon RW. Sequential roles for red blood cell binding proteins enable phased commitment to invasion for malaria parasites. Nat Commun 2023; 14:4619. [PMID: 37528099 PMCID: PMC10393984 DOI: 10.1038/s41467-023-40357-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 07/20/2023] [Indexed: 08/03/2023] Open
Abstract
Invasion of red blood cells (RBCs) by Plasmodium merozoites is critical to their continued survival within the host. Two major protein families, the Duffy binding-like proteins (DBPs/EBAs) and the reticulocyte binding like proteins (RBLs/RHs) have been studied extensively in P. falciparum and are hypothesized to have overlapping, but critical roles just prior to host cell entry. The zoonotic malaria parasite, P. knowlesi, has larger invasive merozoites and contains a smaller, less redundant, DBP and RBL repertoire than P. falciparum. One DBP (DBPα) and one RBL, normocyte binding protein Xa (NBPXa) are essential for invasion of human RBCs. Taking advantage of the unique biological features of P. knowlesi and iterative CRISPR-Cas9 genome editing, we determine the precise order of key invasion milestones and demonstrate distinct roles for each family. These distinct roles support a mechanism for phased commitment to invasion and can be targeted synergistically with invasion inhibitory antibodies.
Collapse
Affiliation(s)
- Melissa N Hart
- Department of Infection Biology, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hawkshead Lane, Hatfield, AL9 7TA, UK
| | - Franziska Mohring
- Department of Infection Biology, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Sophia M DonVito
- Department of Infection Biology, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - James A Thomas
- Department of Infection Biology, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | | | - Gavin J Wright
- Wellcome Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, Wentworth Way, York, YO10 5DD, UK
| | - Ellen Knuepfer
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hawkshead Lane, Hatfield, AL9 7TA, UK
- Malaria Parasitology Laboratory, Francis Crick Institute, London, NW1 1AT, UK
| | - Helen R Saibil
- ISMB, Biological Sciences, Birkbeck, University of London, Malet St, London, WC1E 7HX, UK
| | - Robert W Moon
- Department of Infection Biology, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK.
| |
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: 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
|
19
|
Moskovitz R, Pholcharee T, DonVito SM, Guloglu B, Lowe E, Mohring F, Moon RW, Higgins MK. Structural basis for DARC binding in reticulocyte invasion by Plasmodium vivax. Nat Commun 2023; 14:3637. [PMID: 37336887 DOI: 10.1038/s41467-023-39357-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023] Open
Abstract
The symptoms of malaria occur during the blood stage of infection, when the parasite replicates within human red blood cells. The human malaria parasite, Plasmodium vivax, selectively invades reticulocytes in a process which requires an interaction between the ectodomain of the human DARC receptor and the Plasmodium vivax Duffy-binding protein, PvDBP. Previous studies have revealed that a small helical peptide from DARC binds to region II of PvDBP (PvDBP-RII). However, it is also known that sulphation of tyrosine residues on DARC affects its binding to PvDBP and these residues were not observed in previous structures. We therefore present the structure of PvDBP-RII bound to sulphated DARC peptide, showing that a sulphate on tyrosine 41 binds to a charged pocket on PvDBP-RII. We use molecular dynamics simulations, affinity measurements and growth-inhibition experiments in parasites to confirm the importance of this interaction. We also reveal the epitope for vaccine-elicited growth-inhibitory antibody DB1. This provides a complete understanding of the binding of PvDBP-RII to DARC and will guide the design of vaccines and therapeutics to target this essential interaction.
Collapse
Affiliation(s)
- Re'em Moskovitz
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Tossapol Pholcharee
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Sophia M DonVito
- London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Bora Guloglu
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Edward Lowe
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Franziska Mohring
- London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Robert W Moon
- London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Matthew K Higgins
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK.
| |
Collapse
|
20
|
Dash R, Skillman KM, Pereira L, Mascarenhas A, Dass S, Walke J, Almeida A, Fernandes M, Gomes E, White J, Chery-Karschney L, Khandeparkar A, Rathod PK, Duraisingh MT, Kanjee U. Development of a Plasmodium vivax biobank for functional ex vivo assays. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.17.533128. [PMID: 36993272 PMCID: PMC10055260 DOI: 10.1101/2023.03.17.533128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Background Plasmodium vivax is the second most prevalent cause of malaria yet remains challenging to study due to the lack of a continuous in vitro culture system, highlighting the need to establish a biobank of clinical isolates with multiple freezes per sample for use in functional assays. Different methods for cryopreserving parasite isolates were compared and subsequently the most promising one was validated. Enrichment of early- and late-stage parasites and parasite maturation were quantified to facilitate assay planning. Methods In order to compare cryopreservation protocols, nine clinical P. vivax isolates were frozen with four glycerolyte-based mixtures. Parasite recovery post thaw, post KCl-Percoll enrichment and in short-term in vitro culture was measured via slide microscopy. Enrichment of late-stage parasites by magnetic activated cell sorting (MACS) was measured. Short and long-term storage of parasites at either -80°C or liquid nitrogen were also compared. Results Of the four cryopreservation mixtures, one mixture (glycerolyte:serum:RBC at a 2.5:1.5:1 ratio) resulted in improved parasite recovery and statistically significant (P<0.05) enhancement in parasite survival in short-term in vitro culture. A parasite biobank was subsequently generated using this protocol resulting in a collection with 106 clinical isolates, each with 8 vials. The quality of the biobank was validated by measuring several factors from 47 thaws: the average reduction in parasitemia post-thaw (25.3%); the average fold enrichment post KCl-Percoll (6.65-fold); and the average percent recovery of parasites (22.0%, measured from 30 isolates). During short-term in vitro culture, robust maturation of ring stage parasites to later stages (>20% trophozoites, schizonts and gametocytes) was observed in 60.0% of isolates by 48 hours. Enrichment of mature parasite stages via MACS showed good reproducibility, with an average 30.0% post-MACS parasitemia and an average 5.30 × 10 5 parasites/vial. Finally, the effect of storage temperature was tested, and no large impacts from short-term (7 day) or long term (7 - 10 year) storage at -80°C on parasite recovery, enrichment or viability was observed. Conclusions Here, an optimized freezing method for P. vivax clinical isolates is demonstrated as a template for the generation and validation of a parasite biobank for use in functional assays.
Collapse
|
21
|
Watson QD, Carias LL, Malachin A, Redinger KR, Bosch J, Bardelli M, Moon RW, Draper SJ, Zimmerman PA, King CL. Naturally-acquired and Vaccine-induced Human Monoclonal Antibodies to Plasmodium vivax Duffy Binding Protein Inhibit Invasion of Plasmodium knowlesi (PvDBPOR) Transgenic Parasites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531647. [PMID: 36945444 PMCID: PMC10028882 DOI: 10.1101/2023.03.07.531647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
The Duffy antigen receptor for chemokines (DARC) expressed on erythrocytes is central to Plasmodium vivax (Pv) invasion of reticulocytes. Pv expresses a Duffy binding protein (PvDBP) on merozoites, a DARC ligand, and their protein-protein interaction is central to vivax blood stage malaria. Here we compared the functional activity of humAbs derived from naturally exposed and vaccinated individuals for the first time using easily cultured P. knowlesi (Pk) that had been genetically modified to replace its endogenous PkDBP orthologue with PvDBP to create a transgenic parasite, PkPvDBPOR. This transgenic parasite requires DARC to invade human erythrocytes but is not reticulocyte restricted. Using this model, we evaluated the invasion inhibition potential of 12 humAbs (9 naturally acquired; 3 vaccine-induced) targeting PvDBP individually and in combinations using growth inhibition assays (GIAs). The PvDBP-specific humAbs demonstrated 70-100% inhibition of PkPvDBPOR invasion with the IC50 values ranging from 51 to 338 μg/mL for the 9 naturally acquired (NA) humAbs and 33 to 99 μg/ml for the 3 vaccine-induced (VI) humAbs. To evaluate antagonistic, additive, or synergistic effects, six pairwise combinations were performed using select humAbs. Of these combinations tested, one NA/NA (099100/094083) combination demonstrated relatively strong additive inhibition between 10-100 μg/mL; all combinations of NA and VI humAbs showed additive inhibition at concentrations below 25 μg/mL and antagonism at higher concentrations. None of the humAb combinations showed synergy. This PkPvDBPOR model system enables efficient assessment of NA and VI humAbs individually and in combination.
Collapse
Affiliation(s)
- Quentin D. Watson
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Lenore L. Carias
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Alyssa Malachin
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Karli R. Redinger
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jürgen Bosch
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | - Robert W. Moon
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Simon J. Draper
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Peter A. Zimmerman
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Christopher L. King
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Veterans Affairs Medical Center, Cleveland, OH
| |
Collapse
|
22
|
da Veiga GTS, Moriggi MR, Vettorazzi JF, Müller-Santos M, Albrecht L. Plasmodium vivax vaccine: What is the best way to go? Front Immunol 2023; 13:910236. [PMID: 36726991 PMCID: PMC9885200 DOI: 10.3389/fimmu.2022.910236] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 12/23/2022] [Indexed: 01/18/2023] Open
Abstract
Malaria is one of the most devastating human infectious diseases caused by Plasmodium spp. parasites. A search for an effective and safe vaccine is the main challenge for its eradication. Plasmodium vivax is the second most prevalent Plasmodium species and the most geographically distributed parasite and has been neglected for decades. This has a massive gap in knowledge and consequently in the development of vaccines. The most significant difficulties in obtaining a vaccine against P. vivax are the high genetic diversity and the extremely complex life cycle. Due to its complexity, studies have evaluated P. vivax antigens from different stages as potential targets for an effective vaccine. Therefore, the main vaccine candidates are grouped into preerythrocytic stage vaccines, blood-stage vaccines, and transmission-blocking vaccines. This review aims to support future investigations by presenting the main findings of vivax malaria vaccines to date. There are only a few P. vivax vaccines in clinical trials, and thus far, the best protective efficacy was a vaccine formulated with synthetic peptide from a circumsporozoite protein and Montanide ISA-51 as an adjuvant with 54.5% efficacy in a phase IIa study. In addition, the majority of P. vivax antigen candidates are polymorphic, induce strain-specific and heterogeneous immunity and provide only partial protection. Nevertheless, immunization with recombinant proteins and multiantigen vaccines have shown promising results and have emerged as excellent strategies. However, more studies are necessary to assess the ideal vaccine combination and test it in clinical trials. Developing a safe and effective vaccine against vivax malaria is essential for controlling and eliminating the disease. Therefore, it is necessary to determine what is already known to propose and identify new candidates.
Collapse
Affiliation(s)
- Gisele Tatiane Soares da Veiga
- Laboratory of Apicomplexan Parasites Research, Carlos Chagas Institute, Oswaldo Cruz Foundation (FIOCRUZ), Curitiba, Brazil,Nitrogen Fixation Laboratory, Department of Biochemistry and Molecular Biology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | | | | | - Marcelo Müller-Santos
- Nitrogen Fixation Laboratory, Department of Biochemistry and Molecular Biology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Letusa Albrecht
- Laboratory of Apicomplexan Parasites Research, Carlos Chagas Institute, Oswaldo Cruz Foundation (FIOCRUZ), Curitiba, Brazil,*Correspondence: Letusa Albrecht,
| |
Collapse
|
23
|
Chandley P, Ranjan R, Kumar S, Rohatgi S. Host-parasite interactions during Plasmodium infection: Implications for immunotherapies. Front Immunol 2023; 13:1091961. [PMID: 36685595 PMCID: PMC9845897 DOI: 10.3389/fimmu.2022.1091961] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Malaria is a global infectious disease that remains a leading cause of morbidity and mortality in the developing world. Multiple environmental and host and parasite factors govern the clinical outcomes of malaria. The host immune response against the Plasmodium parasite is heterogenous and stage-specific both in the human host and mosquito vector. The Plasmodium parasite virulence is predominantly associated with its ability to evade the host's immune response. Despite the availability of drug-based therapies, Plasmodium parasites can acquire drug resistance due to high antigenic variations and allelic polymorphisms. The lack of licensed vaccines against Plasmodium infection necessitates the development of effective, safe and successful therapeutics. To design an effective vaccine, it is important to study the immune evasion strategies and stage-specific Plasmodium proteins, which are targets of the host immune response. This review provides an overview of the host immune defense mechanisms and parasite immune evasion strategies during Plasmodium infection. Furthermore, we also summarize and discuss the current progress in various anti-malarial vaccine approaches, along with antibody-based therapy involving monoclonal antibodies, and research advancements in host-directed therapy, which can together open new avenues for developing novel immunotherapies against malaria infection and transmission.
Collapse
Affiliation(s)
- Pankaj Chandley
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Ravikant Ranjan
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Soma Rohatgi
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India,*Correspondence: Soma Rohatgi,
| |
Collapse
|
24
|
Tobin AR, Crow R, Urusova DV, Klima JC, Tolia NH, Strauch E. Inhibition of a malaria host-pathogen interaction by a computationally designed inhibitor. Protein Sci 2023; 32:e4507. [PMID: 36367441 PMCID: PMC9793980 DOI: 10.1002/pro.4507] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/03/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022]
Abstract
Malaria is a substantial global health burden with 229 million cases in 2019 and 450,000 deaths annually. Plasmodium vivax is the most widespread malaria-causing parasite putting 2.5 billion people at risk of infection. P. vivax has a dormant liver stage and therefore can exist for long periods undetected. Its blood-stage can cause severe reactions and hospitalization. Few treatment and detection options are available for this pathogen. A unique characteristic of P. vivax is that it depends on the Duffy antigen/receptor for chemokines (DARC) on the surface of host red blood cells for invasion. P. vivax employs the Duffy binding protein (DBP) to bind to DARC. We first de novo designed a three helical bundle scaffolding database which was screened via protease digestions for stability. Protease-resistant scaffolds highlighted thresholds for stability, which we utilized for selecting DARC mimetics that we subsequentially designed through grafting and redesign of these scaffolds. The optimized design small helical protein disrupts the DBP:DARC interaction. The inhibitor blocks the receptor binding site on DBP and thus forms a strong foundation for a therapeutic that will inhibit reticulocyte infection and prevent the pathogenesis of P. vivax malaria.
Collapse
Affiliation(s)
- Autumn R. Tobin
- Department of Pharmaceutical and Biomedical SciencesUniversity of GeorgiaAthensGeorgiaUSA
| | - Rachel Crow
- Department of MicrobiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Darya V. Urusova
- Department of Molecular MicrobiologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Jason C. Klima
- Institute for Protein DesignUniversity of WashingtonSeattleWashingtonUSA
- Department of BiochemistryUniversity of WashingtonSeattleWashingtonUSA
| | - Niraj H. Tolia
- Department of Molecular MicrobiologyWashington University School of MedicineSaint LouisMissouriUSA
- Host‐Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria, Immunology and Vaccinology, National Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| | - Eva‐Maria Strauch
- Department of Pharmaceutical and Biomedical SciencesUniversity of GeorgiaAthensGeorgiaUSA
- Institute of BioinformaticsUniversity of GeorgiaAthensGeorgiaUSA
| |
Collapse
|
25
|
Mohring F, van Schalkwyk DA, Henrici RC, Blasco B, Leroy D, Sutherland CJ, Moon RW. Cation ATPase (ATP4) Orthologue Replacement in the Malaria Parasite Plasmodium knowlesi Reveals Species-Specific Responses to ATP4-Targeting Drugs. mBio 2022; 13:e0117822. [PMID: 36190127 PMCID: PMC9600963 DOI: 10.1128/mbio.01178-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/31/2022] [Indexed: 11/30/2022] Open
Abstract
Several unrelated classes of antimalarial compounds developed against Plasmodium falciparum target a parasite-specific P-type ATP-dependent Na+ pump, PfATP4. We have previously shown that other malaria parasite species infecting humans are less susceptible to these compounds. Here, we generated a series of transgenic Plasmodium knowlesi orthologue replacement (OR) lines in which the endogenous pkatp4 locus was replaced by a recodonized P. knowlesi atp4 (pkatp4) coding region or the orthologous coding region from P. falciparum, Plasmodium malariae, Plasmodium ovale subsp. curtisi, or Plasmodium vivax. Each OR transgenic line displayed a similar growth pattern to the parental P. knowlesi line. We found significant orthologue-specific differences in parasite susceptibility to three chemically unrelated ATP4 inhibitors, but not to comparator drugs, among the P. knowlesi OR lines. The PfATP4OR transgenic line of P. knowlesi was significantly more susceptible than our control PkATP4OR line to three ATP4 inhibitors: cipargamin, PA21A092, and SJ733. The PvATP4OR and PmATP4OR lines were similarly susceptible to the control PkATP4OR line, but the PocATP4OR line was significantly less susceptible to all ATP4 inhibitors than the PkATP4OR line. Cipargamin-induced inhibition of Na+ efflux was also significantly greater with the P. falciparum orthologue of ATP4. This confirms that species-specific susceptibility differences previously observed in ex vivo studies of human isolates are partly or wholly enshrined in the primary amino acid sequences of the respective ATP4 orthologues and highlights the need to monitor efficacy of investigational malaria drugs against multiple species. P. knowlesi is now established as an important in vitro model for studying drug susceptibility in non-falciparum malaria parasites. IMPORTANCE Effective drugs are vital to minimize the illness and death caused by malaria. Development of new drugs becomes ever more urgent as drug resistance emerges. Among promising compounds now being developed to treat malaria are several unrelated molecules that each inhibit the same protein in the malaria parasite-ATP4. Here, we exploited the genetic tractability of P. knowlesi to replace its own ATP4 genes with orthologues from five human-infective species to understand the drug susceptibility differences among these parasites. We previously estimated the susceptibility to ATP4-targeting drugs of each species using clinical samples from malaria patients. These estimates closely matched those of the corresponding "hybrid" P. knowlesi parasites carrying introduced ATP4 genes. Thus, species-specific ATP4 inhibitor efficacy is directly determined by the sequence of the gene. Our novel approach to understanding cross-species susceptibility/resistance can strongly support the effort to develop antimalarials that effectively target all human malaria parasite species.
Collapse
Affiliation(s)
- Franziska Mohring
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Donelly A. van Schalkwyk
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ryan C. Henrici
- Center for Global Health, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Didier Leroy
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Colin J. Sutherland
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- UK Health Security Agency Malaria Reference Laboratory, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Robert W. Moon
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
26
|
Neutralizing and interfering human antibodies define the structural and mechanistic basis for antigenic diversion. Nat Commun 2022; 13:5888. [PMID: 36202833 PMCID: PMC9537153 DOI: 10.1038/s41467-022-33336-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 09/13/2022] [Indexed: 11/28/2022] Open
Abstract
Defining mechanisms of pathogen immune evasion and neutralization are critical to develop potent vaccines and therapies. Merozoite Surface Protein 1 (MSP-1) is a malaria vaccine antigen and antibodies to MSP-1 are associated with protection from disease. However, MSP-1-based vaccines performed poorly in clinical trials in part due to a limited understanding of the protective antibody response to MSP-1 and of immune evasion by antigenic diversion. Antigenic diversion was identified as a mechanism wherein parasite neutralization by a MSP-1-specific rodent antibody was disrupted by MSP-1-specific non-inhibitory blocking/interfering antibodies. Here, we investigated a panel of MSP-1-specific naturally acquired human monoclonal antibodies (hmAbs). Structures of multiple hmAbs with diverse neutralizing potential in complex with MSP-1 revealed the epitope of a potent strain-transcending hmAb. This neutralizing epitope overlaps with the epitopes of high-affinity non-neutralizing hmAbs. Strikingly, the non-neutralizing hmAbs outcompete the neutralizing hmAb enabling parasite survival. These findings demonstrate the structural and mechanistic basis for a generalizable pathogen immune evasion mechanism through neutralizing and interfering human antibodies elicited by antigenic diversion, and provides insights required to develop potent and durable malaria interventions. The Plasmodium falciparum Merozoite Surface Protein 1 (MSP-1) is a prime vaccine candidate for malaria. Here, the authors structurally and functionally characterise a panel of naturally acquired MSP-1 specific antibodies to identify one with potent broadly neutralising activity and better understand immune evasion mechanisms.
Collapse
|
27
|
Kar S, Sinha A. Plasmodium vivax Duffy Binding Protein-Based Vaccine: a Distant Dream. Front Cell Infect Microbiol 2022; 12:916702. [PMID: 35909975 PMCID: PMC9325973 DOI: 10.3389/fcimb.2022.916702] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
The neglected but highly prevalent Plasmodium vivax in South-east Asia and South America poses a great challenge, with regards to long-term in-vitro culturing and heavily limited functional assays. Such visible challenges as well as narrowed progress in development of experimental research tools hinders development of new drugs and vaccines. The leading vaccine candidate antigen Plasmodium vivax Duffy Binding Protein (PvDBP), is essential for reticulocyte invasion by binding to its cognate receptor, the Duffy Antigen Receptor for Chemokines (DARC), on the host’s reticulocyte surface. Despite its highly polymorphic nature, the amino-terminal cysteine-rich region II of PvDBP (PvDBPII) has been considered as an attractive target for vaccine-mediated immunity and has successfully completed the clinical trial Phase 1. Although this molecule is an attractive vaccine candidate against vivax malaria, there is still a question on its viability due to recent findings, suggesting that there are still some aspects which needs to be looked into further. The highly polymorphic nature of PvDBPII and strain-specific immunity due to PvDBPII allelic variation in Bc epitopes may complicate vaccine efficacy. Emergence of various blood-stage antigens, such as PvRBP, PvEBP and supposedly many more might stand in the way of attaining full protection from PvDBPII. As a result, there is an urgent need to assess and re-assess various caveats connected to PvDBP, which might help in designing a long-term promising vaccine for P. vivax malaria. This review mainly deals with a bunch of rising concerns for validation of DBPII as a vaccine candidate antigen for P. vivax malaria.
Collapse
|
28
|
Thiam LG, Mangou K, Ba A, Mbengue A, Bei AK. Leveraging genome editing to functionally evaluate Plasmodium diversity. Trends Parasitol 2022; 38:558-571. [PMID: 35469746 DOI: 10.1016/j.pt.2022.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 12/13/2022]
Abstract
The ambitious goal of malaria elimination requires an in-depth understanding of the parasite's biology to counter the growing threat of antimalarial resistance and immune evasion. Timely assessment of the functional impact of antigenic diversity in the early stages of vaccine development will be critical for achieving the goal of malaria control, elimination, and ultimately eradication. Recent advances in targeted genome editing enabled the functional validation of resistance-associated markers in Plasmodium falciparum, the deadliest malaria-causing pathogen and strain-specific immune neutralization. This review explores recent advances made in leveraging genome editing to aid the functional evaluation of Plasmodium diversity and highlights how these techniques can assist in prioritizing both therapeutic and vaccine candidates.
Collapse
Affiliation(s)
- Laty Gaye Thiam
- G4 - Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Khadidiatou Mangou
- G4 - Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Aboubacar Ba
- G4 - Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, 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
| | - 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 06510, USA.
| |
Collapse
|
29
|
Moehrle JJ. Development of New Strategies for Malaria Chemoprophylaxis: From Monoclonal Antibodies to Long-Acting Injectable Drugs. Trop Med Infect Dis 2022; 7:tropicalmed7040058. [PMID: 35448833 PMCID: PMC9024890 DOI: 10.3390/tropicalmed7040058] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 02/04/2023] Open
Abstract
Drug discovery for malaria has traditionally focused on orally available drugs that kill the abundant, parasitic blood stage. Recently, there has also been an interest in injectable medicines, in the form of monoclonal antibodies (mAbs) with long-lasting plasma half-lives or long-lasting depot formulations of small molecules. These could act as prophylactic drugs, targeting the sporozoites and other earlier parasitic stages in the liver, when the parasites are less numerous, or as another intervention strategy targeting the formation of infectious gametocytes. Generally speaking, the development of mAbs is less risky (costly) than small-molecule drugs, and they have an excellent safety profile with few or no off-target effects. Therefore, populations who are the most vulnerable to malaria, i.e., pregnant women and young children would have access to such new treatments much faster than is presently the case for new antimalarials. An analysis of mAbs that were successfully developed for oncology illustrates some of the feasibility aspects, and their potential as affordable drugs in low- and middle-income countries.
Collapse
Affiliation(s)
- Joerg J Moehrle
- Integrated Sciences, R&D, Medicines for Malaria Venture, Route de Pré Bois 20, CH-1215 Geneva 15, Switzerland
| |
Collapse
|
30
|
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
|
31
|
Alves JRS, de Araújo FF, Pires CV, Teixeira-Carvalho A, Lima BAS, Torres LM, Ntumngia FB, Adams JH, Kano FS, Carvalho LH. Multiplexed Microsphere-Based Flow Cytometric Assay to Assess Strain Transcending Antibodies to Plasmodium vivax Duffy Binding Protein II Reveals an Efficient Tool to Identify Binding-Inhibitory Antibody Responders. Front Immunol 2021; 12:704653. [PMID: 34675915 PMCID: PMC8523986 DOI: 10.3389/fimmu.2021.704653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 09/13/2021] [Indexed: 11/24/2022] Open
Abstract
Malaria remains a major public health problem worldwide, and Plasmodium vivax is the most widely distributed malaria parasite. Naturally acquired binding inhibitory antibodies (BIAbs) to region II of the Duffy binding protein (DBPII), a P. vivax ligand that is critical for reticulocyte invasion, are associated with a reduced risk of clinical malaria. Owing to methodological issues in evaluating antibodies that inhibit the DBPII-DARC interaction, a limited number of studies have investigated DBPII BIAbs in P. vivax-exposed populations. Based on the assumption that individuals with a consistent BIAb response are characterized by strain-transcending immune responses, we hypothesized that detecting broadly reactive DBPII antibodies would indicate the presence of BIAb response. By taking advantage of an engineered DBPII immunogen targeting conserved DBPII neutralizing epitopes (DEKnull-2), we standardized a multiplex flow cytometry-based serological assay to detect broadly neutralizing IgG antibodies. For this study, a standard in vitro cytoadherence assay with COS-7 cells expressing DBPII was used to test for DBPII BIAb response in long-term P. vivax-exposed Amazonian individuals. Taken together, the results demonstrate that this DBPII-based multiplex assay facilitates identifying DBPII BIAb carriers. Of relevance, the ability of the multiplex assay to identify BIAb responders was highly accurate when the positivity for all antigens was considered. In conclusion, the standardized DBPII-based flow cytometric assay confirmed that DBPII-BIAb activity was associated with the breadth rather than the magnitude of anti-DBPII antibodies. Altogether, our results suggest that multiplex detection of broadly DBPII-reactive antibodies facilitates preliminary screening of BIAb responders.
Collapse
Affiliation(s)
- Jéssica R. S. Alves
- Molecular Biology and Malaria Immunology, René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ), Belo Horizonte, Brazil
| | - Fernanda F. de Araújo
- Integrated Research Group in Biomarkers, René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ), Belo Horizonte, Brazil
| | - Camilla V. Pires
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, FL, United States
| | - Andréa Teixeira-Carvalho
- Integrated Research Group in Biomarkers, René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ), Belo Horizonte, Brazil
| | - Barbara A. S. Lima
- Molecular Biology and Malaria Immunology, René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ), Belo Horizonte, Brazil
| | - Letícia M. Torres
- Molecular Biology and Malaria Immunology, René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ), Belo Horizonte, Brazil
| | - Francis B. Ntumngia
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, FL, United States
| | - John H. Adams
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, FL, United States
| | - Flora S. Kano
- Molecular Biology and Malaria Immunology, René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ), Belo Horizonte, Brazil
| | - Luzia H. Carvalho
- Molecular Biology and Malaria Immunology, René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ), Belo Horizonte, Brazil
| |
Collapse
|
32
|
Baird JK. Basic Research of Plasmodium vivax Biology Enabling Its Management as a Clinical and Public Health Problem. Front Cell Infect Microbiol 2021; 11:696598. [PMID: 34540716 PMCID: PMC8447957 DOI: 10.3389/fcimb.2021.696598] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/22/2021] [Indexed: 12/20/2022] Open
Abstract
The emerging understanding of Plasmodium vivax as an infection seated in extravascular spaces of its human host carries fundamentally important implications for its management as a complex clinical and public health problem. This progress begins to reverse decades of neglected research borne of the false dogma of P. vivax as an intrinsically benign and inconsequential parasite. This Review provides real world context for the on-going laboratory explorations of the molecular and cellular events in the life of this parasite. Chemotherapies against the latent reservoir impose extraordinarily complex and difficult problems of science and medicine, but great strides in studies of the biology of hepatic P. vivax promise solutions. Fundamental assumptions regarding the interpretation of parasitaemia in epidemiology, clinical medicine, and public health are being revisited and reassessed in light of new studies of P. vivax cellular/molecular biology and pathogenesis. By examining these long overlooked complexities of P. vivax malaria, we open multiple new avenues to vaccination, chemoprevention, countermeasures against transmission, epidemiology, diagnosis, chemotherapy, and clinical management. This Review expresses how clarity of vision of biology and pathogenesis may rationally and radically transform the multiple means by which we may combat this insidiously harmful infection.
Collapse
Affiliation(s)
- J Kevin Baird
- Eijkman-Oxford Clinical Research Unit, Eijkman Institute of Molecular Biology, Jakarta, Indonesia.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
33
|
Opi DH, Kurtovic L, Chan JA, Horton JL, Feng G, Beeson JG. Multi-functional antibody profiling for malaria vaccine development and evaluation. Expert Rev Vaccines 2021; 20:1257-1272. [PMID: 34530671 DOI: 10.1080/14760584.2021.1981864] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION A vaccine would greatly accelerate current global efforts toward malaria elimination. While a partially efficacious vaccine has been achieved for Plasmodium falciparum, a major bottleneck in developing highly efficacious vaccines is a lack of reliable correlates of protection, and the limited application of assays that quantify functional immune responses to evaluate and down-select vaccine candidates in pre-clinical studies and clinical trials. AREAS COVERED In this review, we describe the important role of antibodies in immunity against malaria and detail the nature and functional activities of antibodies against the malaria-causing parasite. We highlight the growing understanding of antibody effector functions against malaria and in vitro assays to measure these functional antibody responses. We discuss the application of these assays to quantify antibody functions in vaccine development and evaluation. EXPERT OPINION It is becoming increasingly clear that multiple antibody effector functions are involved in immunity to malaria. Therefore, we propose that evaluating vaccine candidates needs to move beyond individual assays or measuring IgG magnitude alone. Instead, vaccine evaluation should incorporate the full breadth of antibody response types and harness a wider range of assays measuring functional antibody responses. We propose a 3-tier approach to implementing assays to inform vaccine evaluation.
Collapse
Affiliation(s)
- D Herbert Opi
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Liriye Kurtovic
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia
| | - Jo-Anne Chan
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Jessica L Horton
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Gaoqian Feng
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - James G Beeson
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia.,Department of Microbiology, Monash University, Clayton, Australia
| |
Collapse
|
34
|
Ong JJY, Russell B, Han JH. Plasmodium cynomolgi Berok Growth Inhibition Assay by Thiol-reactive Probe Based Flow Cytometric Measurement. Bio Protoc 2021; 11:e4147. [PMID: 34604452 DOI: 10.21769/bioprotoc.4147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 01/18/2023] Open
Abstract
The relapsing malaria species, Plasmodium vivax, is the most widely distributed and difficult-to-treat cause of human malaria. The merozoites of P. vivax preferentially invade ephemeral human CD71+ reticulocytes (nascent reticulocytes), thereby limiting the development of a robust continuous culture in vitro. Fortunately, P. vivax's sister species, P. cynomolgi Berok, can be cultured continuously, providing the ability to screen novel therapeutics drug and vaccine candidates in a reliable and high-throughput manner. Based on well-established growth inhibition activity (GIA) assays against P. falciparum and P. knowlesi, this protocol adopts the current flow cytometry assay methodology and investigates P. vivax inhibitory antibodies using the P. cynomolgi Berok invasion model based on the thiol-reactivity and DNA abundance of viable parasites in macaque erythrocytes. Established GIA assays screen antibodies at either a single concentration or high/low dose concentrations to provide quick insights for prioritizing potential antibodies capable of specifically interrupting parasite ligand and host receptor binding with minimal concentrations. Hence, this protocol expands on the existing GIA assay by using serially diluted antibodies and generating a dose-response curve to better quantify the inhibitory efficacy amongst selected vaccine candidates.
Collapse
Affiliation(s)
- Jessica Jie Ying Ong
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Bruce Russell
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Jin-Hee Han
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.,Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| |
Collapse
|
35
|
Golassa L, Messele A, Oriero EC, Amambua-Ngwa A. Sequence analysis of Plasmodium vivax Duffy binding proteins reveals the presence of unique haplotypes and diversifying selection in Ethiopian isolates. Malar J 2021; 20:312. [PMID: 34246262 PMCID: PMC8271342 DOI: 10.1186/s12936-021-03843-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/04/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Red blood cell invasion by the Plasmodium vivax merozoite requires interaction between the Duffy antigen receptor for chemokines (DARC) and the P. vivax Duffy-binding protein II (PvDBPII). Given that the disruption of this interaction prevents P. vivax blood-stage infection, a PvDBP-based vaccine development has been well recognized. However, the polymorphic nature of PvDBPII prevents a strain transcending immune response and complicates attempts to design a vaccine. METHODS Twenty-three P. vivax clinical isolates collected from three areas of Ethiopia were sequenced at the pvdbpII locus. A total of 392 global pvdbpII sequences from seven P. vivax endemic countries were also retrieved from the NCBI archive for comparative analysis of genetic diversity, departure from neutrality, linkage disequilibrium, genetic differentiation, PvDBP polymorphisms, recombination and population structure of the parasite population. To establish a haplotype relationship a network was constructed using the median joining algorithm. RESULTS A total of 110 variable sites were found, of which 44 were parsimony informative. For Ethiopian isolates there were 12 variable sites of which 10 were parsimony informative. These parsimony informative variants resulted in 10 nonsynonymous mutations. The overall haplotype diversity for global isolates was 0.9596; however, the haplotype diversity was 0.874 for Ethiopia. Fst values for genetic revealed Ethiopian isolates were closest to Indian isolates as well as to Sri Lankan and Sudanese isolates but further away from Mexican, Papua New Guinean and South Korean isolates. There was a total of 136 haplotypes from the 415 global isolates included for this study. Haplotype prevalence ranged from 36.76% to 0.7%, from this 74.2% were represented by single parasite isolates. None of the Ethiopian isolates grouped with the Sal I reference haplotype. From the total observed nonsynonymous mutations 13 mapped to experimentally verified epitope sequences. Including 10 non-synonymous mutations from Ethiopia. However, all the polymorphic regions in Ethiopian isolates were located away from DARC, responsible for junction formation. CONCLUSION The results of this study are concurrent with the multivalent vaccine approach to design an effective treatment. However, the presence of novel haplotypes in Ethiopian isolates that were not shared by other global sequences warrant further investigation.
Collapse
Affiliation(s)
- Lemu Golassa
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia.
| | - Alebachew Messele
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Eniyou Cheryll Oriero
- Medical Research Council Unit The Gambia At London, School of Hygiene and Tropical Medicin, Banjul, The Gambia
| | - Alfred Amambua-Ngwa
- Medical Research Council Unit The Gambia At London, School of Hygiene and Tropical Medicin, Banjul, The Gambia
| |
Collapse
|
36
|
Ndegwa DN, Kundu P, Hostetler JB, Marin-Menendez A, Sanderson T, Mwikali K, Verzier LH, Coyle R, Adjalley S, Rayner JC. Using Plasmodium knowlesi as a model for screening Plasmodium vivax blood-stage malaria vaccine targets reveals new candidates. PLoS Pathog 2021; 17:e1008864. [PMID: 34197567 PMCID: PMC8279373 DOI: 10.1371/journal.ppat.1008864] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 07/14/2021] [Accepted: 06/01/2021] [Indexed: 11/18/2022] Open
Abstract
Plasmodium vivax is responsible for the majority of malaria cases outside Africa. Unlike P. falciparum, the P. vivax life-cycle includes a dormant liver stage, the hypnozoite, which can cause infection in the absence of mosquito transmission. An effective vaccine against P. vivax blood stages would limit symptoms and pathology from such recurrent infections, and therefore could play a critical role in the control of this species. Vaccine development in P. vivax, however, lags considerably behind P. falciparum, which has many identified targets with several having transitioned to Phase II testing. By contrast only one P. vivax blood-stage vaccine candidate based on the Duffy Binding Protein (PvDBP), has reached Phase Ia, in large part because the lack of a continuous in vitro culture system for P. vivax limits systematic screening of new candidates. We used the close phylogenetic relationship between P. vivax and P. knowlesi, for which an in vitro culture system in human erythrocytes exists, to test the scalability of systematic reverse vaccinology to identify and prioritise P. vivax blood-stage targets. A panel of P. vivax proteins predicted to function in erythrocyte invasion were expressed as full-length recombinant ectodomains in a mammalian expression system. Eight of these antigens were used to generate polyclonal antibodies, which were screened for their ability to recognize orthologous proteins in P. knowlesi. These antibodies were then tested for inhibition of growth and invasion of both wild type P. knowlesi and chimeric P. knowlesi lines modified using CRISPR/Cas9 to exchange P. knowlesi genes with their P. vivax orthologues. Candidates that induced antibodies that inhibited invasion to a similar level as PvDBP were identified, confirming the utility of P. knowlesi as a model for P. vivax vaccine development and prioritizing antigens for further follow up.
Collapse
Affiliation(s)
- Duncan N. Ndegwa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Department of Biological Sciences, University of Embu, Embu, Kenya
| | - Prasun Kundu
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, United Kingdom
| | - Jessica B. Hostetler
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | | | - Theo Sanderson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Kioko Mwikali
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Lisa H. Verzier
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Rachael Coyle
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Sophie Adjalley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Julian C. Rayner
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, United Kingdom
| |
Collapse
|
37
|
De SL, May S, Shah K, Slawinski M, Changrob S, Xu S, Barnes SJ, Chootong P, Ntumngia FB, Adams JH. Variable immunogenicity of a vivax malaria blood-stage vaccine candidate. Vaccine 2021; 39:2668-2675. [PMID: 33840564 DOI: 10.1016/j.vaccine.2021.03.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 02/25/2021] [Accepted: 03/20/2021] [Indexed: 10/21/2022]
Abstract
Relapsing malaria caused by Plasmodium vivax is a neglected tropical disease and an important cause of malaria worldwide. Vaccines to prevent clinical disease and mosquito transmission of vivax malaria are needed to overcome the distinct challenges of this important public health problem. In this vaccine immunogenicity study in mice, we examined key variables of responses to a P. vivax Duffy binding protein vaccine, a leading candidate to prevent the disease-causing blood-stages. Significant sex-dependent differences were observed in B cell (CD80+) and T cell (CD8+) central memory subsets, resulting in significant differences in functional immunogenicity and durability of anti-DBP protective efficacy. These significant sex-dependent differences in inbred mice were in the CD73+CD80+ memory B cell, H2KhiCD38hi/lo, and effector memory subsets. This study highlights sex and immune genes as critical variables that can impact host responses to P. vivax antigens and must be taken into consideration when designing clinical vaccine studies.
Collapse
Affiliation(s)
- Sai Lata De
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL 33612, United States
| | - Samuel May
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL 33612, United States
| | - Keshav Shah
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL 33612, United States
| | - Michelle Slawinski
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL 33612, United States
| | - Siriruk Changrob
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Shulin Xu
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL 33612, United States
| | - Samantha J Barnes
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL 33612, United States
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Francis B Ntumngia
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL 33612, United States.
| | - John H Adams
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL 33612, United States.
| |
Collapse
|
38
|
Kaslow DC. Malaria vaccine research & innovation: the intersection of IA2030 and zero malaria. NPJ Vaccines 2020; 5:109. [PMID: 33298967 PMCID: PMC7677906 DOI: 10.1038/s41541-020-00259-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 10/30/2020] [Indexed: 12/17/2022] Open
Affiliation(s)
- David C Kaslow
- PATH, 2201 Westlake Avenue, Suite 200, Seattle, WA, 98121, USA.
| |
Collapse
|
39
|
Mittal P, Mishra S, Kar S, Pande V, Sinha A, Sharma A. Global distribution of single amino acid polymorphisms in Plasmodium vivax Duffy-binding-like domain and implications for vaccine development efforts. Open Biol 2020; 10:200180. [PMID: 32993415 PMCID: PMC7536081 DOI: 10.1098/rsob.200180] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Plasmodium vivax (Pv) malaria continues to be geographically widespread with approximately 15 million worldwide cases annually. Along with other proteins, Duffy-binding proteins (DBPs) are used by plasmodium for RBC invasion and the parasite-encoded receptor binding regions lie in their Duffy-binding-like (DBL) domains-thus making it a prime vaccine candidate. This study explores the sequence diversity in PvDBL globally, with an emphasis on India as it remains a major contributor to the global Pv malaria burden. Based on 1358 PvDBL protein sequences available in NCBI, we identified 140 polymorphic sites within 315 residues of PvDBL. Alarmingly, country-wise mapping of SAAPs from field isolates revealed varied and distinct polymorphic profiles for different nations. We report here 31 polymorphic residue positions in the global SAAP profile, most of which map to the PvDBL subdomain 2 (α1-α6). A distinct clustering of SAAPs distal to the DARC-binding sites is indicative of immune evasive strategies by the parasite. Analyses of PvDBL-neutralizing antibody complexes revealed that between 24% and 54% of interface residues are polymorphic. This work provides a framework to recce and expand the polymorphic space coverage in PvDBLs as this has direct implications for vaccine development studies. It also emphasizes the significance of surveying global SAAP distributions before or alongside the identification of vaccine candidates.
Collapse
Affiliation(s)
- Payal Mittal
- Molecular Medicine Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India.,ICMR-National Institute of Malaria Research, New Delhi, 110077, India
| | - Siddhartha Mishra
- Molecular Medicine Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India.,ICMR-National Institute of Malaria Research, New Delhi, 110077, India
| | - Sonalika Kar
- ICMR-National Institute of Malaria Research, New Delhi, 110077, India.,Department of Biotechnology, Kumaun University, Nainital, Uttarakhand, 263001 India
| | - Veena Pande
- Department of Biotechnology, Kumaun University, Nainital, Uttarakhand, 263001 India
| | - Abhinav Sinha
- ICMR-National Institute of Malaria Research, New Delhi, 110077, India
| | - Amit Sharma
- Molecular Medicine Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India.,ICMR-National Institute of Malaria Research, New Delhi, 110077, India
| |
Collapse
|
40
|
Kanjee U, Grüring C, Babar P, Meyers A, Dash R, Pereira L, Mascarenhas A, Chaand M, Rangel GW, Clark MA, Chery L, Gomes E, Rathod PK, Duraisingh MT. Plasmodium vivax Strains Use Alternative Pathways for Invasion. J Infect Dis 2020; 223:1817-1821. [PMID: 32941614 DOI: 10.1093/infdis/jiaa592] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/16/2020] [Indexed: 12/19/2022] Open
Abstract
Plasmodium vivax has 2 invasion ligand/host receptor pathways (P. vivax Duffy-binding protein/Duffy antigen receptor for chemokines [DARC] and P. vivax reticulocyte binding protein 2b/transferrin receptor [TfR1]) that are promising targets for therapeutic intervention. We optimized invasion assays with isogenic cultured reticulocytes. Using a receptor blockade approach with multiple P. vivax isolates, we found that all strains utilized both DARC and TfR1, but with significant variation in receptor usage. This suggests that P. vivax, like Plasmodium falciparum, uses alternative invasion pathways, with implications for pathogenesis and vaccine development.
Collapse
Affiliation(s)
- Usheer Kanjee
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Christof Grüring
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Prasad Babar
- University of Washington, Seattle, Washington, USA.,Department of Medicine, Goa Medical College Hospital, Bambolim, Goa, India
| | - Anosha Meyers
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Rashmi Dash
- University of Washington, Seattle, Washington, USA.,Department of Medicine, Goa Medical College Hospital, Bambolim, Goa, India
| | - Ligia Pereira
- University of Washington, Seattle, Washington, USA.,Department of Medicine, Goa Medical College Hospital, Bambolim, Goa, India
| | - Anjali Mascarenhas
- University of Washington, Seattle, Washington, USA.,Department of Medicine, Goa Medical College Hospital, Bambolim, Goa, India
| | - Mudit Chaand
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Gabriel W Rangel
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Martha A Clark
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Laura Chery
- University of Washington, Seattle, Washington, USA
| | - Edwin Gomes
- Department of Medicine, Goa Medical College Hospital, Bambolim, Goa, India
| | | | | |
Collapse
|
41
|
Schäfer C, Roobsoong W, Kangwanrangsan N, Bardelli M, Rawlinson TA, Dambrauskas N, Trakhimets O, Parthiban C, Goswami D, Reynolds LM, Kennedy SY, Flannery EL, Murphy SC, Sather DN, Draper SJ, Sattabongkot J, Mikolajczak SA, Kappe SHI. A Humanized Mouse Model for Plasmodium vivax to Test Interventions that Block Liver Stage to Blood Stage Transition and Blood Stage Infection. iScience 2020; 23:101381. [PMID: 32739836 PMCID: PMC7399188 DOI: 10.1016/j.isci.2020.101381] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/02/2020] [Accepted: 07/15/2020] [Indexed: 12/20/2022] Open
Abstract
The human malaria parasite Plasmodium vivax remains vastly understudied, mainly due to the lack of suitable laboratory models. Here, we report a humanized mouse model to test interventions that block P. vivax parasite transition from liver stage infection to blood stage infection. Human liver-chimeric FRGN huHep mice infected with P. vivax sporozoites were infused with human reticulocytes, allowing transition of exo-erythrocytic merozoites to reticulocyte infection and development into all erythrocytic forms, including gametocytes, in vivo. In order to test the utility of this model for preclinical assessment of interventions, the invasion blocking potential of a monoclonal antibody targeting the essential interaction of the P. vivax Duffy Binding Protein with the Duffy antigen receptor was tested by passive immunization. This antibody inhibited invasion by over 95%, providing unprecedented in vivo evidence that PvDBP constitutes a promising blood stage vaccine candidate and proving our model highly suitable to test blood stage interventions.
Collapse
Affiliation(s)
- Carola Schäfer
- 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 10400, Thailand
| | - Niwat Kangwanrangsan
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | | | | | - Nicholas Dambrauskas
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Olesya Trakhimets
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Chaitra Parthiban
- Departments of Laboratory Medicine and Microbiology and Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - Debashree Goswami
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Laura M Reynolds
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Spencer Y Kennedy
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Erika L Flannery
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Sean C Murphy
- Departments of Laboratory Medicine and Microbiology and Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - D Noah Sather
- 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; Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - 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; Department of Global Health, University of Washington, Seattle, WA 98105, USA.
| |
Collapse
|
42
|
Hotspots in Plasmodium and RBC Receptor-Ligand Interactions: Key Pieces for Inhibiting Malarial Parasite Invasion. Int J Mol Sci 2020; 21:ijms21134729. [PMID: 32630804 PMCID: PMC7370042 DOI: 10.3390/ijms21134729] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/15/2020] [Accepted: 05/24/2020] [Indexed: 11/17/2022] Open
Abstract
Protein-protein interactions (IPP) play an essential role in practically all biological processes, including those related to microorganism invasion of their host cells. It has been found that a broad repertoire of receptor-ligand interactions takes place in the binding interphase with host cells in malaria, these being vital interactions for successful parasite invasion. Several trials have been conducted for elucidating the molecular interface of interactions between some Plasmodium falciparum and Plasmodium vivax antigens with receptors on erythrocytes and/or reticulocytes. Structural information concerning these complexes is available; however, deeper analysis is required for correlating structural, functional (binding, invasion, and inhibition), and polymorphism data for elucidating new interaction hotspots to which malaria control methods can be directed. This review describes and discusses recent structural and functional details regarding three relevant interactions during erythrocyte invasion: Duffy-binding protein 1 (DBP1)–Duffy antigen receptor for chemokines (DARC); reticulocyte-binding protein homolog 5 (PfRh5)-basigin, and erythrocyte binding antigen 175 (EBA175)-glycophorin A (GPA).
Collapse
|
43
|
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: 152] [Impact Index Per Article: 30.4] [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
|
44
|
Abstract
Much of the gain in malaria control, in terms of regional achievements in restricting geographical spread and reducing malaria cases and deaths, can be attributed to large-scale deployment of antimalarial drugs, insecticide-treated bed nets, and early diagnostics. However, despite impressive progress, control efforts have stalled because of logistics, unsustainable delivery, or short-term effectiveness of existing interventions or a combination of these reasons. A highly efficacious malaria vaccine as an additional tool would go a long way, but success in the development of this important intervention remains elusive. Moreover, most of the vaccine candidate antigens that were investigated in early-stage clinical trials, selected partly because of their immunogenicity and abundance during natural malaria infection, were polymorphic or structurally complex or both. Likewise, we have a limited understanding of immune mechanisms that confer protection. We reflect on some considerable technological and scientific progress that has been achieved and the lessons learned.
Collapse
Affiliation(s)
- Nirianne Marie Q Palacpac
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Toshihiro Horii
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| |
Collapse
|
45
|
Aitken EH, Mahanty S, Rogerson SJ. Antibody effector functions in malaria and other parasitic diseases: a few needles and many haystacks. Immunol Cell Biol 2020; 98:264-275. [DOI: 10.1111/imcb.12320] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/02/2020] [Accepted: 01/28/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Elizabeth H Aitken
- Department of Medicine The Doherty Institute The University of Melbourne 792 Elizabeth Street Melbourne VIC 3000 Australia
| | - Siddhartha Mahanty
- Department of Medicine The Doherty Institute The University of Melbourne 792 Elizabeth Street Melbourne VIC 3000 Australia
| | - Stephen J Rogerson
- Department of Medicine The Doherty Institute The University of Melbourne 792 Elizabeth Street Melbourne VIC 3000 Australia
| |
Collapse
|
46
|
Mohring F, Hart MN, Patel A, Baker DA, Moon RW. CRISPR-Cas9 Genome Editing of Plasmodium knowlesi. Bio Protoc 2020; 10:e3522. [PMID: 33654746 PMCID: PMC7842605 DOI: 10.21769/bioprotoc.3522] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/15/2019] [Accepted: 12/23/2019] [Indexed: 12/28/2022] Open
Abstract
Plasmodium knowlesi is a zoonotic malaria parasite in Southeast Asia that can cause severe and fatal malaria in humans. The main hosts are Macaques, but modern diagnostic tools reveal increasing numbers of human infections. After P. falciparum, P. knowlesi is the only other malaria parasite capable of being maintained in long term in vitro culture with human red blood cells (RBCs). Its closer ancestry to other non-falciparum human malaria parasites, more balanced AT-content, larger merozoites and higher transfection efficiencies, gives P. knowlesi some key advantages over P. falciparum for the study of malaria parasite cell/molecular biology. Here, we describe the generation of marker-free CRISPR gene-edited P. knowlesi parasites, the fast and scalable production of transfection constructs and analysis of transfection efficiencies. Our protocol allows rapid, reliable and unlimited rounds of genome editing in P. knowlesi requiring only a single recyclable selection marker.
Collapse
Affiliation(s)
- Franziska Mohring
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Melissa N. Hart
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Avnish Patel
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, United Kingdom
| | - David A. Baker
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Robert W. Moon
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, United Kingdom
| |
Collapse
|
47
|
Chan L, Dietrich MH, Nguitragool W, Tham W. Plasmodium vivax Reticulocyte Binding Proteins for invasion into reticulocytes. Cell Microbiol 2020; 22:e13110. [PMID: 31469946 PMCID: PMC7003471 DOI: 10.1111/cmi.13110] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/25/2019] [Accepted: 08/25/2019] [Indexed: 12/22/2022]
Abstract
Plasmodium vivax is responsible for most of the malaria infections outside Africa and is currently the predominant malaria parasite in countries under elimination programs. P. vivax preferentially enters young red cells called reticulocytes. Advances in understanding the molecular and cellular mechanisms of entry are hampered by the inability to grow large numbers of P. vivax parasites in a long-term in vitro culture. Recent progress in understanding the biology of the P. vivax Reticulocyte Binding Protein (PvRBPs) family of invasion ligands has led to the identification of a new invasion pathway into reticulocytes, an understanding of their structural architecture and PvRBPs as targets of the protective immune response to P. vivax infection. This review summarises current knowledge on the role of reticulocytes in P. vivax infection, the function of the PvRBP family of proteins in generating an immune response in human populations, and the characterization of anti-PvRBP antibodies in blocking parasite invasion.
Collapse
Affiliation(s)
- Li‐Jin Chan
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- Department of Medical BiologyThe University of MelbourneMelbourneVictoriaAustralia
| | - Melanie H. Dietrich
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- Department of Medical BiologyThe University of MelbourneMelbourneVictoriaAustralia
| | - Wang Nguitragool
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical MedicineMahidol UniversityBangkokThailand
| | - Wai‐Hong Tham
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- Department of Medical BiologyThe University of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
48
|
Good MF, Stanisic DI. Whole parasite vaccines for the asexual blood stages ofPlasmodium. Immunol Rev 2019; 293:270-282. [DOI: 10.1111/imr.12819] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 08/27/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Michael F. Good
- Institute for Glycomics Griffith University Gold Coast Qld. Australia
| | | |
Collapse
|
49
|
Duffy PE. Structure Solves the Problem with Malaria Merozoite Vaccines. Trends Parasitol 2019; 35:855-857. [PMID: 31623952 PMCID: PMC11131090 DOI: 10.1016/j.pt.2019.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 09/16/2019] [Indexed: 12/14/2022]
Abstract
Malaria vaccines targeting merozoite invasion of erythrocytes have long held appeal but failed in clinical trials. Three structural studies of antibody-antigen complexes by Alanine et al., Urusova et al., and Rawlinson et al. define neutralizing and nonneutralizing epitopes in essential invasion proteins, leading to rational design of improved merozoite vaccines.
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.
| |
Collapse
|
50
|
Antonelli LR, Junqueira C, Vinetz JM, Golenbock DT, Ferreira MU, Gazzinelli RT. The immunology of Plasmodium vivax malaria. Immunol Rev 2019; 293:163-189. [PMID: 31642531 DOI: 10.1111/imr.12816] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 09/10/2019] [Indexed: 12/13/2022]
Abstract
Plasmodium vivax infection, the predominant cause of malaria in Asia and Latin America, affects ~14 million individuals annually, with considerable adverse effects on wellbeing and socioeconomic development. A clinical hallmark of Plasmodium infection, the paroxysm, is driven by pyrogenic cytokines produced during the immune response. Here, we review studies on the role of specific immune cell types, cognate innate immune receptors, and inflammatory cytokines on parasite control and disease symptoms. This review also summarizes studies on recurrent infections in individuals living in endemic regions as well as asymptomatic infections, a serious barrier to eliminating this disease. We propose potential mechanisms behind these repeated and subclinical infections, such as poor induction of immunological memory cells and inefficient T effector cells. We address the role of antibody-mediated resistance to P. vivax infection and discuss current progress in vaccine development. Finally, we review immunoregulatory mechanisms, such as inhibitory receptors, T regulatory cells, and the anti-inflammatory cytokine, IL-10, that antagonizes both innate and acquired immune responses, interfering with the development of protective immunity and parasite clearance. These studies provide new insights for the clinical management of symptomatic as well as asymptomatic individuals and the development of an efficacious vaccine for vivax malaria.
Collapse
Affiliation(s)
- Lis R Antonelli
- Instituto de Pesquisas Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Caroline Junqueira
- Instituto de Pesquisas Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Joseph M Vinetz
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Douglas T Golenbock
- Division of Infectious Disease and immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Marcelo U Ferreira
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Ricardo T Gazzinelli
- Instituto de Pesquisas Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil.,Division of Infectious Disease and immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.,Plataforma de Medicina Translacional, Fundação Oswaldo Cruz, Ribeirão Preto, Brazil
| |
Collapse
|