1
|
Eisa M, Flores N, Khedr O, Gomez-Escobar E, Bédard N, Abdeltawab NF, Bruneau J, Grakoui A, Shoukry NH. Activation-Induced Marker Assay to Identify and Isolate HCV-Specific T Cells for Single-Cell RNA-Seq Analysis. Viruses 2024; 16:1623. [PMID: 39459954 PMCID: PMC11512294 DOI: 10.3390/v16101623] [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: 08/05/2024] [Revised: 10/01/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Identification and isolation of antigen-specific T cells for downstream transcriptomic analysis is key for various immunological studies. Traditional methods using major histocompatibility complex (MHC) multimers are limited by the number of predefined immunodominant epitopes and MHC matching of the study subjects. Activation-induced markers (AIM) enable highly sensitive detection of rare antigen-specific T cells irrespective of the availability of MHC multimers. Herein, we have developed an AIM assay for the detection, sorting and subsequent single-cell RNA sequencing (scRNA-seq) analysis of hepatitis C virus (HCV)-specific T cells. We examined different combinations of the activation markers CD69, CD40L, OX40, and 4-1BB at 6, 9, 18 and 24 h post stimulation with HCV peptide pools. AIM+ CD4 T cells exhibited upregulation of CD69 and CD40L as early as 6 h post-stimulation, while OX40 and 4-1BB expression was delayed until 18 h. AIM+ CD8 T cells were characterized by the coexpression of CD69 and 4-1BB at 18 h, while the expression of CD40L and OX40 remained low throughout the stimulation period. AIM+ CD4 and CD8 T cells were successfully sorted and processed for scRNA-seq analysis examining gene expression and T cell receptor (TCR) usage. scRNA-seq analysis from this one subject revealed that AIM+ CD4 T (CD69+ CD40L+) cells predominantly represented Tfh, Th1, and Th17 profiles, whereas AIM+ CD8 T (CD69+ 4-1BB+) cells primarily exhibited effector and effector memory profiles. TCR analysis identified 1023 and 160 unique clonotypes within AIM+ CD4 and CD8 T cells, respectively. In conclusion, this approach offers highly sensitive detection of HCV-specific T cells that can be applied for cohort studies, thus facilitating the identification of specific gene signatures associated with infection outcome and vaccination.
Collapse
Affiliation(s)
- Mohamed Eisa
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Tour Viger, Local R09.414, 900 rue St-Denis, Montréal, QC H2X 0A9, Canada (N.F.A.)
| | - Nicol Flores
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Tour Viger, Local R09.414, 900 rue St-Denis, Montréal, QC H2X 0A9, Canada (N.F.A.)
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Omar Khedr
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Tour Viger, Local R09.414, 900 rue St-Denis, Montréal, QC H2X 0A9, Canada (N.F.A.)
| | - Elsa Gomez-Escobar
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Tour Viger, Local R09.414, 900 rue St-Denis, Montréal, QC H2X 0A9, Canada (N.F.A.)
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Nathalie Bédard
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Tour Viger, Local R09.414, 900 rue St-Denis, Montréal, QC H2X 0A9, Canada (N.F.A.)
| | - Nourtan F. Abdeltawab
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Tour Viger, Local R09.414, 900 rue St-Denis, Montréal, QC H2X 0A9, Canada (N.F.A.)
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo 3296121, Egypt
- School of Pharmacy, Newgiza University, Giza 3296121, Egypt
| | - Julie Bruneau
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Tour Viger, Local R09.414, 900 rue St-Denis, Montréal, QC H2X 0A9, Canada (N.F.A.)
- Département de Médecine Familiale et Département d’Urgence, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Arash Grakoui
- Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Naglaa H. Shoukry
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Tour Viger, Local R09.414, 900 rue St-Denis, Montréal, QC H2X 0A9, Canada (N.F.A.)
- Département de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| |
Collapse
|
2
|
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
|
3
|
Miura K. How to Accelerate Early Stage of Malaria Vaccine Development by Optimizing Functional Assays. Vaccines (Basel) 2024; 12:586. [PMID: 38932315 PMCID: PMC11209467 DOI: 10.3390/vaccines12060586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
While two Plasmodium falciparum circumsporozoite protein-based pre-erythrocytic vaccines (PEV), RTS,S and R21, have been approved by the WHO, no blood-stage vaccine (BSV) or transmission-blocking vaccine (TBV) has reached a phase 3 trial. One of the major obstacles that slows down malaria vaccine development is the shortage (or lack) of in vitro assays or animal models by which investigators can reasonably select the best vaccine formulation (e.g., antigen, adjuvant, or platform) and/or immunization strategy (e.g., interval of inoculation or route of immunization) before a human phase 2 trial. In the case of PEV, RTS,S and R21 have set a benchmark, and a new vaccine can be compared with (one of) the approved PEV directly in preclinical or early clinical studies. However, such an approach cannot be utilized for BSV or TBV development at this moment. The focus of this review is in vitro assays or in vivo models that can be used for P. falciparum BSV or TBV development, and I discuss important considerations during assay selection, standardization, qualification, validation, and interpretation of the assay results. Establishment of a robust assay/model with proper interpretation of the results is the one of key elements to accelerate future vaccine development.
Collapse
Affiliation(s)
- Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
4
|
Borkens Y. Malaria & mRNA Vaccines: A Possible Salvation from One of the Most Relevant Infectious Diseases of the Global South. Acta Parasitol 2023; 68:916-928. [PMID: 37828249 PMCID: PMC10665248 DOI: 10.1007/s11686-023-00712-y] [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: 10/10/2022] [Accepted: 08/01/2023] [Indexed: 10/14/2023]
Abstract
Malaria is one of the most dangerous infectious diseases in the world. It occurs in tropical and subtropical regions and affects about 40% of the world´s population. In endemic regions, an estimated 200 million people contract malaria each year. Three-quarters of all global deaths (about 600 per year) are children under 5 years of age. Thus, malaria is one of the most relevant tropical and also childhood diseases in the world. Thanks to various public health measures such as vector control through mosquito nets or the targeted use of insecticides as well as the use of antimalarial prophylaxis drugs, the incidence has already been successfully reduced in recent years. However, to reduce the risk of malaria and to protect children effectively, further measures are necessary. An important part of these measures is an effective vaccination against malaria. However, the history of research shows that the development of an effective malaria vaccine is not an easy undertaking and is associated with some complications. Research into possible vaccines began as early as the 1960s. However, the results achieved were rather sobering and the various vaccines fell short of their expectations. It was not until 2015 that the vaccine RTS,S/AS01 received a positive evaluation from the European Medicines Agency. Since then, the vaccine has been tested in Africa. However, with the COVID-19 pandemic, there are new developments in vaccine research that could also benefit malaria research. These include, among others, the so-called mRNA vaccines. Already in the early 1990s, an immune response triggered by an mRNA vaccine was described for the first time. Since then, mRNA vaccines have been researched and discussed for possible prophylaxis. However, it was not until the COVID-19 pandemic that these vaccines experienced a veritable progress. mRNA vaccines against SARS-CoV-2 were rapidly developed and achieved high efficacy in studies. Based on this success, it is not surprising that companies are also focusing on other diseases and pathogens. Besides viral diseases, such as influenza or AIDS, malaria is high on this list. Many pharmaceutical companies (including the German companies BioNTech and CureVac) have already confirmed that they are researching mRNA vaccines against malaria. However, this is not an easy task. The aim of this article is to describe and discuss possible antigens that could be considered for mRNA vaccination. However, this topic is currently still very speculative.
Collapse
Affiliation(s)
- Yannick Borkens
- Charité, Charitéplatz 1, 10117, Berlin, Germany.
- Humboldt-Universität zu Berlin, Unter den Linden 6, 10117, Berlin, Germany.
| |
Collapse
|
5
|
Poloni C, Schonhofer C, Ivison S, Levings MK, Steiner TS, Cook L. T-cell activation-induced marker assays in health and disease. Immunol Cell Biol 2023; 101:491-503. [PMID: 36825901 PMCID: PMC10952637 DOI: 10.1111/imcb.12636] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/18/2023] [Accepted: 02/22/2023] [Indexed: 02/25/2023]
Abstract
Activation-induced marker (AIM) assays have proven to be an accessible and rapid means of antigen-specific T-cell detection. The method typically involves short-term incubation of whole blood or peripheral blood mononuclear cells with antigens of interest, where autologous antigen-presenting cells process and present peptides in complex with major histocompatibility complex (MHC) molecules. Recognition of peptide-MHC complexes by T-cell receptors then induces upregulation of activation markers on the T cells that can be detected by flow cytometry. In this review, we highlight the most widely used activation markers for assays in the literature while identifying nuances and potential downfalls associated with the technique. We provide a summary of how AIM assays have been used in both discovery science and clinical studies, including studies of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunity. This review primarily focuses on AIM assays using human blood or peripheral blood mononuclear cell samples, with some considerations noted for tissue-derived T cells and nonhuman samples. AIM assays are a powerful tool that enables detailed analysis of antigen-specific T-cell frequency, phenotype and function without needing to know the precise antigenic peptides and their MHC restriction elements, enabling a wider analysis of immunity generated following infection and/or vaccination.
Collapse
Affiliation(s)
- Chad Poloni
- Division of Infectious Diseases, Department of MedicineUniversity of British ColumbiaVancouverBCCanada
- BC Children's Hospital Research InstituteVancouverBCCanada
| | - Cole Schonhofer
- Division of Infectious Diseases, Department of MedicineUniversity of British ColumbiaVancouverBCCanada
- BC Children's Hospital Research InstituteVancouverBCCanada
| | - Sabine Ivison
- BC Children's Hospital Research InstituteVancouverBCCanada
- Department of SurgeryUniversity of British ColumbiaVancouverBCCanada
| | - Megan K Levings
- BC Children's Hospital Research InstituteVancouverBCCanada
- Department of SurgeryUniversity of British ColumbiaVancouverBCCanada
| | - Theodore S Steiner
- Division of Infectious Diseases, Department of MedicineUniversity of British ColumbiaVancouverBCCanada
- BC Children's Hospital Research InstituteVancouverBCCanada
| | - Laura Cook
- Division of Infectious Diseases, Department of MedicineUniversity of British ColumbiaVancouverBCCanada
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
- Department of Critical Care, Melbourne Medical SchoolUniversity of MelbourneMelbourneAustralia
| |
Collapse
|
6
|
Structural analysis of Plasmodium falciparum ookinete surface antigen Pfs28 relevant for malaria vaccine design. Sci Rep 2022; 12:19556. [PMID: 36379968 PMCID: PMC9664031 DOI: 10.1038/s41598-022-24054-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Pfs28 is a Plasmodium falciparum malaria transmission-blocking vaccine candidate that is anchored to the parasite surface through a C-terminal glycosylphosphatidylinositol (GPI) moiety, and plays a role in parasite survival in the mosquito midgut. Pfs28 contains epidermal growth factor (EGF)-like domains and is part of a family of sexual stage malaria proteins that includes the related vaccine antigen Pfs25. The lack of structural definition of Pfs28 and the immune response to this candidate has limited further malaria vaccine development for this antigen. Here, we present the crystal structure of Pfs28, examine its conservation with P. vivax Pvs28, and evaluate the cross-reactivity of Pfs28 to antibodies that recognize Pfs25. Pfs28 is comprised of four EGF-like domains stabilized by ten disulfide bridges with an overall architecture that highly resembles Pfs25. Despite the high sequence and structural similarity between these antigens, no cross reactivity of Pfs28 to anti-Pfs25 monoclonal antibodies could be demonstrated.
Collapse
|
7
|
Iyori M, Blagborough AM, Mizuno T, Abe YI, Nagaoka M, Hori N, Yamagoshi I, Da DF, Gregory WF, Hasyim AA, Yamamoto Y, Sakamoto A, Yoshida K, Mizukami H, Shida H, Yoshida S. Sterile protection and transmission blockade by a multistage anti-malarial vaccine in the pre-clinical study. Front Immunol 2022; 13:1005476. [PMID: 36248835 PMCID: PMC9558734 DOI: 10.3389/fimmu.2022.1005476] [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: 07/28/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
The Malaria Vaccine Technology Roadmap 2013 (World Health Organization) aims to develop safe and effective vaccines by 2030 that will offer at least 75% protective efficacy against clinical malaria and reduce parasite transmission. Here, we demonstrate a highly effective multistage vaccine against both the pre-erythrocytic and sexual stages of Plasmodium falciparum that protects and reduces transmission in a murine model. The vaccine is based on a viral-vectored vaccine platform, comprising a highly-attenuated vaccinia virus strain, LC16m8Δ (m8Δ), a genetically stable variant of a licensed and highly effective Japanese smallpox vaccine LC16m8, and an adeno-associated virus (AAV), a viral vector for human gene therapy. The genes encoding P. falciparum circumsporozoite protein (PfCSP) and the ookinete protein P25 (Pfs25) are expressed as a Pfs25-PfCSP fusion protein, and the heterologous m8Δ-prime/AAV-boost immunization regimen in mice provided both 100% protection against PfCSP-transgenic P. berghei sporozoites and up to 100% transmission blocking efficacy, as determined by a direct membrane feeding assay using parasites from P. falciparum-positive, naturally-infected donors from endemic settings. Remarkably, the persistence of vaccine-induced immune responses were over 7 months and additionally provided complete protection against repeated parasite challenge in a murine model. We propose that application of the m8Δ/AAV malaria multistage vaccine platform has the potential to contribute to the landmark goals of the malaria vaccine technology roadmap, to achieve life-long sterile protection and high-level transmission blocking efficacy.
Collapse
Affiliation(s)
- Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | | | - Tetsushi Mizuno
- Department of Parasitology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Yu-ichi Abe
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | - Mio Nagaoka
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | - Naoto Hori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | - Iroha Yamagoshi
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | - Dari F. Da
- Département de Biologie Médicale et Santé Publique, Unité Paludisme et Maladies Tropicales Négligées, Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - William F. Gregory
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Ammar A. Hasyim
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | - Yutaro Yamamoto
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | - Akihiko Sakamoto
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | - Kunitaka Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | - Hiroaki Mizukami
- Division of Gene Therapy, Jichi Medical University, Tochigi, Japan
| | - Hisatoshi Shida
- Institute for Genetic Medicine, Hokkaido University, Hokkaido, Japan
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| |
Collapse
|
8
|
Borkens Y. Malaria-Antigene in der Ära der mRNA-Impfstoffe. Monatsschr Kinderheilkd 2022; 170:828-838. [PMID: 35855690 PMCID: PMC9281189 DOI: 10.1007/s00112-022-01554-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 04/25/2022] [Indexed: 11/28/2022]
Abstract
ZusammenfassungBereits in den frühen 1990er-Jahren wurde erstmals eine durch einen mRNA-Impfstoff ausgelöste Immunantwort beschrieben. Seitdem wurden mRNA-Impfstoffe für eine mögliche Prophylaxe erforscht und diskutiert. Doch erst mit der COVID-19-Pandemie erlebten diese Impfstoffe einen wahren Boom. Die ersten mRNA-Impfstoffe wurden gegen SARS-CoV‑2 zugelassen und zeigten große Erfolge. Es ist daher nicht verwunderlich, dass sich die Hersteller auch auf andere Krankheiten und Pathogene konzentrieren. Neben viralen Krankheiten wie Influenza oder Aids steht Malaria weit oben auf dieser Liste. Viele Pharmaunternehmen (u. a. die deutschen Unternehmen BioNTech und CureVac) haben bereits bestätigt, an mRNA-Impfstoffen gegen Malaria zu forschen. Dabei ist die Entwicklung eines funktionierenden Impfstoffes gegen Malaria kein leichtes Unterfangen. Seit den 1960ern wird an möglichen Impfstoffen geforscht. Die Ergebnisse sind dabei eher ernüchternd. Erst 2015 erhielt der Impfstoff RTS,S/AS01 eine positive Bewertung der Europäischen Arzneimittel-Agentur. Seitdem wird der Impfstoff in Afrika getestet.
Collapse
Affiliation(s)
- Yannick Borkens
- College of Public Health, Medical and Veterinary Science, James Cook University, 1 James Cook Drive, 4811 Townsville, Queensland Australien
| |
Collapse
|
9
|
Kaslow DC. Efforts to Develop Pfs25 Vaccines. Am J Trop Med Hyg 2022; 107:tpmd211326. [PMID: 35895392 DOI: 10.4269/ajtmh.21-1326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/10/2022] [Indexed: 02/18/2024] Open
Abstract
Acknowledging the fallibilities of recalling events from more than three decades ago, the recollection of Richard Carter's impact on the identification and development of Pfs25, a major surface protein of Plasmodium falciparum zygotes and ookinetes, and target of malaria transmission-blocking vaccines, remains unassailable. In fondest memories of Richard Carter's many contributions, herein retells some memorable events along the tortuous journey toward the development of Pfs25 vaccines.
Collapse
|