1
|
Flores-Garcia Y, Salgado-Jimenez B, Park M, Mathis-Torres S, Locke E, MacGill RS, Moskovitz R, Wilson IA, Zavala F. Epitope specificity of antibody-mediated protection induced in mice by the malaria vaccine RTS,S/AS01. NPJ Vaccines 2025; 10:101. [PMID: 40394009 PMCID: PMC12092751 DOI: 10.1038/s41541-025-01162-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 05/12/2025] [Indexed: 05/22/2025] Open
Abstract
Antibodies induced by the malaria vaccine RTS,S/AS01 neutralize infectivity of transgenic sporozoites expressing Plasmodium falciparum CSP (PfCSP). These antibodies recognize the junctional, minor repeats, central repeats, and C-term regions of this antigen. The epitope specificity of antibodies mediating protection in mice was characterized in vivo using transgenic sporozoites expressing restricted antigenic portions of PfCSP. In this model, we found protection is mediated mostly by antibodies specific for the central repeats.
Collapse
Affiliation(s)
- Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Berenice Salgado-Jimenez
- Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Minah Park
- Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Shamika Mathis-Torres
- Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Emily Locke
- Center for Vaccine Innovation and Access, PATH, Washington, DC, USA
| | | | - Re'em Moskovitz
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
2
|
Langowski MD, Francica JR, Roederer AL, Hurlburt NK, Rodarte JV, Da Silva Pereira L, Flynn BJ, Bonilla B, Dillon M, Kiyuka P, Ravichandran R, Weidle C, Carter L, Rao M, Matyas GR, Pepper M, Idris AH, Seder RA, Pancera M, King NP. Elicitation of liver-stage immunity by nanoparticle immunogens displaying P. falciparum CSP-derived antigens. NPJ Vaccines 2025; 10:87. [PMID: 40325041 PMCID: PMC12053698 DOI: 10.1038/s41541-025-01140-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 04/23/2025] [Indexed: 05/07/2025] Open
Abstract
A vaccine that provides robust, durable protection against malaria remains a global health priority. Although a breakthrough in the fight against malaria has recently been achieved by the licensure of two vaccines based on the circumsporozoite protein (CSP), the effectiveness and durability of protection can still be improved. Both vaccines contain a portion of CSP that does not include epitopes targeted by recently identified, potently protective monoclonal antibodies, suggesting that newer immunogens can expand the breadth of immunity and potentially increase protection. Here we explored >100 alternative CSP-based immunogens and evaluated the immunogenicity and protection of a large number of candidates, comparing several to the licensed R21 vaccine. The data highlight several general features that improve the stability and immunogenicity of CSP-based vaccines, such as inclusion of the C-terminal domain and high-density display on protein nanoparticle scaffolds. We also identify antigen design strategies that do not warrant further exploration, such as synthetic repeat regions that include non-native repeat cadences. The benchmark R21 vaccine outperformed our best immunogen for immunogenicity and protection. Overall, our data provide valuable insights on the inclusion of junctional region epitopes that will guide the development of potent and durable vaccines against malaria.
Collapse
Affiliation(s)
- Mark D Langowski
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, WA, USA
| | - Joseph R Francica
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alex L Roederer
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Nicholas K Hurlburt
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Justas V Rodarte
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lais Da Silva Pereira
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barbara J Flynn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Brian Bonilla
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Marlon Dillon
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patience Kiyuka
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rashmi Ravichandran
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Connor Weidle
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lauren Carter
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Mangala Rao
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- U.S. Military HIV Research Program, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Gary R Matyas
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- U.S. Military HIV Research Program, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Marion Pepper
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Azza H Idris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Marie Pancera
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| | - Neil P King
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
| |
Collapse
|
3
|
Krenger PS, Roques M, Vogt ACS, Pardini A, Rothen DA, Balke I, Schnider ST, Mohsen MO, Heussler VT, Zeltins A, Bachmann MF. Probing novel epitopes on the Plasmodium falciparum circumsporozoite protein for vaccine development. NPJ Vaccines 2024; 9:225. [PMID: 39557901 PMCID: PMC11574195 DOI: 10.1038/s41541-024-01006-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 10/22/2024] [Indexed: 11/20/2024] Open
Abstract
RTS,S and R21 are the only vaccines recommended by the WHO to protect children from Plasmodium falciparum (Pf) clinical malaria. Both vaccines target the Pf sporozoite surface protein circumsporozoite protein (CSP). Recent studies showed that human antibodies neutralize Pf sporozoites most efficiently when simultaneously binding to the PfCSP NANP repeat and the NPDP junction domain. However, neither RTS,S nor R21 targets this junction domain. To test the potential of the NPDP junction domain and other sites of PfCSP as innovative vaccine targets, we developed multiple vaccine candidates based on cucumber mosaic virus-like particles (CuMVTT-VLPs). These candidates vary in several aspects: the number of targeted NANP repeats, the presence or absence of the junction domain, the cleavage site, and up to three NVDP repeats within the target sequence. Immunogenicity and efficacy studies were conducted in BALB/c mice, utilizing chimeric Plasmodium berghei (Pb) sporozoites, in which the endogenous CSP has been replaced by PfCSP (Pb/PfCSP). We observed a positive association between the number of targeted NANP repeats and the induction of specific IgM/IgG antibodies. Elevated humoral responses led to enhanced protection against parasitemia after Pb/PfCSP sporozoite challenge. Especially high-avidity/affinity antibody formation and vaccine protection were NANP repeat-dependent. Intriguingly, vaccine efficacy was not enhanced by targeting sites on PfCSP other than the NANP repeats. Our data emphasize the dominant role of the NANP repeat region for induction of protective antibodies. Furthermore, we present here novel malaria vaccine candidates with an excellent immunogenic profile that confer sterile protection in mice, even in absence of adjuvants.
Collapse
Affiliation(s)
- Pascal S Krenger
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
- Department for Rheumatology and Immunology, University Hospital of Bern, Bern, Switzerland.
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland.
| | - Magali Roques
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Anne-Cathrine S Vogt
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department for Rheumatology and Immunology, University Hospital of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
| | - Alessandro Pardini
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department for Rheumatology and Immunology, University Hospital of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
| | - Dominik A Rothen
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department for Rheumatology and Immunology, University Hospital of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
| | - Ina Balke
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Sophie T Schnider
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department for Rheumatology and Immunology, University Hospital of Bern, Bern, Switzerland
| | - Mona O Mohsen
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department for Rheumatology and Immunology, University Hospital of Bern, Bern, Switzerland
| | | | - Andris Zeltins
- Latvian Biomedical Research and Study Centre, Riga, Latvia
- Saiba AG, Pfäffikon, Switzerland
| | - Martin F Bachmann
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
- Department for Rheumatology and Immunology, University Hospital of Bern, Bern, Switzerland.
- Nuffield Department of Medicine, Centre for Cellular and Molecular Physiology (CCMP), The Jenner Institute, University of Oxford, Oxford, UK.
| |
Collapse
|
4
|
Plieskatt J, Ofori EA, Naghizadeh M, Miura K, Flores-Garcia Y, Borbye-Lorenzen N, Tiono AB, Skogstrand K, Sagara I, Zavala F, Theisen M. ProC6C, a novel multi-stage malaria vaccine, elicits functional antibodies against the minor and central repeats of the Circumsporozoite Protein in human adults. Front Immunol 2024; 15:1481829. [PMID: 39555079 PMCID: PMC11563800 DOI: 10.3389/fimmu.2024.1481829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/11/2024] [Indexed: 11/19/2024] Open
Abstract
Introduction ProC6C is a multi-stage malaria vaccine which includes Plasmodium falciparum Circumsporozoite Protein (PfCSP), Pfs48/45 and Pfs230 sequences, designed to elicit functional antibodies that prevent sporozoite invasion of human hepatocytes (PfCSP) and parasite development in mosquitoes (Pfs48/45 and Pfs230). ProC6C formulated on Alhydrogel was evaluated in combination with Matrix-M in a Phase 1 trial in Burkina Faso. The PfCSP antibody responses were assessed for magnitude, specificity, avidity and functionality. These results compliment the prior reported safety and tolerability of ProC6C as well as the transmission reducing activity of ProC6C. Methods The PfCSP response of ProC6C in Burkinabes in the Phase 1 trial (PACTR202201848463189) was profiled through the three vaccine administrations of 100 µg protein on Alhydrogel® alone (AlOH) or combined with 50 µg Matrix-M™ adjuvant (AlOH/Matrix-M). Serology was completed against full-length PfCSP and major/minor repeat peptides using antibody equivalence to PfCSP monoclonal antibodies (mAb 311, mAb 317 and mAb L9). Comparison of the ProC6C responses were made to those that received RTS,S/AS01 in a study conducted in Thailand. Bio-Layer Interferometry was further used to determine antibody avidity. The human IgG was subsequently purified, pooled, and evaluated in a mouse sporozoite challenge model to determine functionality. Results A single administration of ProC6C-AlOH/Matrix-M seroconverted 19 of 20 volunteers against PfCSP and significantly enhanced antibody titers to major and minor repeats (and present through D180). At D70, ProC6C-AlOH/Matrix-M PfCSP antibodies were found to be similar to responder pools generated from Thai adults receiving RTS,S/AS01. Additionally, ProC6C antibodies were found to be competitive to established PfCSP antibodies such as mAb 317 and mAb L9. The purified and pooled IgG from human volunteers, used in a passive transfer mouse sporozoite challenge model, showed a median of 50% inhibition (P=0.0058). ProC6C PfCSP antibodies were functional in this in vivo assessment and consistent with inhibition seen by other Circumsporozoite vaccines in this model. Discussion This analysis supports continued investigation of the antibody responses elicited by the ProC6C multi-stage malaria vaccine. This Phase 1 clinical trial demonstrated the short PfCSP sequence included in ProC6C can induce significant PfCSP antibodies in humans, which importantly were determined to be functional.
Collapse
Affiliation(s)
- Jordan Plieskatt
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
| | - Ebenezer Addo Ofori
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
- Centre for Translational Medicine and Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Mohammad Naghizadeh
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
- Centre for Translational Medicine and Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Nis Borbye-Lorenzen
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
| | - Alfred B. Tiono
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Kristin Skogstrand
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
| | - Issaka Sagara
- Malaria Research and Training Center, Mali- National Institute of Allergy and Infectious Diseases International Center for Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
- Centre for Translational Medicine and Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
5
|
Wang LT, Idris AH, Kisalu NK, Crompton PD, Seder RA. Monoclonal antibodies to the circumsporozoite proteins as an emerging tool for malaria prevention. Nat Immunol 2024; 25:1530-1545. [PMID: 39198635 DOI: 10.1038/s41590-024-01938-2] [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: 05/06/2024] [Accepted: 07/17/2024] [Indexed: 09/01/2024]
Abstract
Despite various public health strategies, malaria caused by Plasmodium falciparum parasites remains a major global health challenge that requires development of new interventions. Extended half-life human monoclonal antibodies targeting the P. falciparum circumsporozoite protein on sporozoites, the infective form of malaria parasites, prevent malaria in rodents and humans and have been advanced into clinical development. The protective epitopes on the circumsporozoite protein targeted by monoclonal antibodies have been defined. Cryogenic electron and multiphoton microscopy have enabled mechanistic structural and functional investigations of how antibodies bind to the circumsporozoite protein and neutralize sporozoites. Moreover, innovations in bioinformatics and antibody engineering have facilitated enhancement of antibody potency and durability. Here, we summarize the latest scientific advances in understanding how monoclonal antibodies to the circumsporozoite protein prevent malaria and highlight existing clinical data and future plans for how this emerging intervention can be used alone or alongside existing antimalarial interventions to control malaria across at-risk populations.
Collapse
Affiliation(s)
- Lawrence T Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Azza H Idris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
| | - Neville K Kisalu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- PATH's Center for Vaccine Innovation and Access, Washington, DC, USA
| | - Peter D Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
6
|
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
|
7
|
Stump WH, Klingenberg HJ, Ott AC, Gonzales DM, Burns JM. Design and Evaluation of Chimeric Plasmodium falciparum Circumsporozoite Protein-Based Malaria Vaccines. Vaccines (Basel) 2024; 12:351. [PMID: 38675734 PMCID: PMC11053680 DOI: 10.3390/vaccines12040351] [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: 02/21/2024] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Efficacy data on two malaria vaccines, RTS,S and R21, targeting Plasmodium falciparum circumsporozoite protein (PfCSP), are encouraging. Efficacy may be improved by induction of additional antibodies to neutralizing epitopes outside of the central immunodominant repeat domain of PfCSP. We designed four rPfCSP-based vaccines in an effort to improve the diversity of the antibody response. We also evaluated P. falciparum merozoite surface protein 8 (PfMSP8) as a malaria-specific carrier protein as an alternative to hepatitis B surface antigen. We measured the magnitude, specificity, subclass, avidity, durability, and efficacy of vaccine-induced antibodies in outbred CD1 mice. In comparison to N-terminal- or C-terminal-focused constructs, immunization with near full-length vaccines, rPfCSP (#1) or the chimeric rPfCSP/8 (#2), markedly increased the breadth of B cell epitopes recognized covering the N-terminal domain, junctional region, and central repeat. Both rPfCSP (#1) and rPfCSP/8 (#2) also elicited a high proportion of antibodies to conformation-dependent epitopes in the C-terminus of PfCSP. Fusion of PfCSP to PfMSP8 shifted the specificity of the T cell response away from PfCSP toward PfMSP8 epitopes. Challenge studies with transgenic Plasmodium yoelii sporozoites expressing PfCSP demonstrated high and consistent sterile protection following rPfCSP/8 (#2) immunization. Of note, antibodies to conformational C-terminal epitopes were not required for protection. These results indicate that inclusion of the N-terminal domain of PfCSP can drive responses to protective, repeat, and non-repeat B cell epitopes and that PfMSP8 is an effective carrier for induction of high-titer, durable anti-PfCSP antibodies.
Collapse
Affiliation(s)
| | | | | | | | - James M. Burns
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA; (W.H.S.); (H.J.K.); (A.C.O.); (D.M.G.)
| |
Collapse
|
8
|
Watson FN, Shears MJ, Kalata AC, Duncombe CJ, Seilie AM, Chavtur C, Conrad E, Cruz Talavera I, Raappana A, Sather DN, Chakravarty S, Sim BKL, Hoffman SL, Tsuji M, Murphy SC. Ultra-low volume intradermal administration of radiation-attenuated sporozoites with the glycolipid adjuvant 7DW8-5 completely protects mice against malaria. Sci Rep 2024; 14:2881. [PMID: 38311678 PMCID: PMC10838921 DOI: 10.1038/s41598-024-53118-9] [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: 01/19/2023] [Accepted: 01/28/2024] [Indexed: 02/06/2024] Open
Abstract
Radiation-attenuated sporozoite (RAS) vaccines can completely prevent blood stage Plasmodium infection by inducing liver-resident memory CD8+ T cells to target parasites in the liver. Such T cells can be induced by 'Prime-and-trap' vaccination, which here combines DNA priming against the P. yoelii circumsporozoite protein (CSP) with a subsequent intravenous (IV) dose of liver-homing RAS to "trap" the activated and expanding T cells in the liver. Prime-and-trap confers durable protection in mice, and efforts are underway to translate this vaccine strategy to the clinic. However, it is unclear whether the RAS trapping dose must be strictly administered by the IV route. Here we show that intradermal (ID) RAS administration can be as effective as IV administration if RAS are co-administrated with the glycolipid adjuvant 7DW8-5 in an ultra-low inoculation volume. In mice, the co-administration of RAS and 7DW8-5 in ultra-low ID volumes (2.5 µL) was completely protective and dose sparing compared to standard volumes (10-50 µL) and induced protective levels of CSP-specific CD8+ T cells in the liver. Our finding that adjuvants and ultra-low volumes are required for ID RAS efficacy may explain why prior reports about higher volumes of unadjuvanted ID RAS proved less effective than IV RAS. The ID route may offer significant translational advantages over the IV route and could improve sporozoite vaccine development.
Collapse
Affiliation(s)
- Felicia N Watson
- Graduate Program in Pathobiology, Department of Global Health, University of Washington, Seattle, WA, 98109, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA
| | - Melanie J Shears
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA
| | - Anya C Kalata
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA
| | - Caroline J Duncombe
- Graduate Program in Pathobiology, Department of Global Health, University of Washington, Seattle, WA, 98109, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA
| | - A Mariko Seilie
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA
| | - Chris Chavtur
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA
| | - Ethan Conrad
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA
| | - Irene Cruz Talavera
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA
| | - Andrew Raappana
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98109, USA
| | - D Noah Sather
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98109, USA
| | - Sumana Chakravarty
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - B Kim Lee Sim
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Stephen L Hoffman
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Sean C Murphy
- Graduate Program in Pathobiology, Department of Global Health, University of Washington, Seattle, WA, 98109, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA.
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA.
- Department of Microbiology, University of Washington, Seattle, WA, 98109, USA.
- Washington National Primate Research Center, University of Washington, Seattle, WA, 98109, USA.
- Department of Laboratories, Seattle Children's Hospital, Seattle, WA, 98105, USA.
| |
Collapse
|
9
|
Williams KL, Guerrero S, Flores-Garcia Y, Kim D, Williamson KS, Siska C, Smidt P, Jepson SZ, Li K, Dennison SM, Mathis-Torres S, Chen X, Wille-Reece U, MacGill RS, Walker M, Jongert E, King CR, Ockenhouse C, Glanville J, Moon JE, Regules JA, Tan YC, Cavet G, Lippow SM, Robinson WH, Dutta S, Tomaras GD, Zavala F, Ketchem RR, Emerling DE. A candidate antibody drug for prevention of malaria. Nat Med 2024; 30:117-129. [PMID: 38167935 PMCID: PMC10803262 DOI: 10.1038/s41591-023-02659-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 10/20/2023] [Indexed: 01/05/2024]
Abstract
Over 75% of malaria-attributable deaths occur in children under the age of 5 years. However, the first malaria vaccine recommended by the World Health Organization (WHO) for pediatric use, RTS,S/AS01 (Mosquirix), has modest efficacy. Complementary strategies, including monoclonal antibodies, will be important in efforts to eradicate malaria. Here we characterize the circulating B cell repertoires of 45 RTS,S/AS01 vaccinees and discover monoclonal antibodies for development as potential therapeutics. We generated >28,000 antibody sequences and tested 481 antibodies for binding activity and 125 antibodies for antimalaria activity in vivo. Through these analyses we identified correlations suggesting that sequences in Plasmodium falciparum circumsporozoite protein, the target antigen in RTS,S/AS01, may induce immunodominant antibody responses that limit more protective, but subdominant, responses. Using binding studies, mouse malaria models, biomanufacturing assessments and protein stability assays, we selected AB-000224 and AB-007088 for advancement as a clinical lead and backup. We engineered the variable domains (Fv) of both antibodies to enable low-cost manufacturing at scale for distribution to pediatric populations, in alignment with WHO's preferred product guidelines. The engineered clone with the optimal manufacturing and drug property profile, MAM01, was advanced into clinical development.
Collapse
Affiliation(s)
| | | | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Dongkyoon Kim
- Atreca, Inc., San Carlos, CA, USA
- Initium Therapeutics, Inc., Natick, MA, USA
| | | | | | | | | | - Kan Li
- Duke Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA
| | - S Moses Dennison
- Duke Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA
| | - Shamika Mathis-Torres
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Ulrike Wille-Reece
- BioNTech US, Inc., Cambridge, MA, USA
- PATH Center for Vaccine Innovation and Access, Washington DC, USA
| | | | | | | | - C Richter King
- PATH Center for Vaccine Innovation and Access, Washington DC, USA
| | | | | | - James E Moon
- Center for Enabling Capabilities, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Jason A Regules
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Yann Chong Tan
- Atreca, Inc., San Carlos, CA, USA
- Nuevocor Pte. Ltd, Singapore, Singapore
| | - Guy Cavet
- Atreca, Inc., San Carlos, CA, USA
- Paramune, Inc., San Carlos, CA, USA
| | | | - William H Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sheetij Dutta
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Georgia D Tomaras
- Duke Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA
- Departments of Immunology, Molecular Genetics and Microbiology, Human Vaccine Institute, Duke University, Durham, NC, USA
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | | |
Collapse
|
10
|
Bolton JS, MacGill RS, Locke E, Regules JA, Bergmann-Leitner ES. Novel antibody competition binding assay identifies distinct serological profiles associated with protection. Front Immunol 2023; 14:1303446. [PMID: 38152401 PMCID: PMC10752609 DOI: 10.3389/fimmu.2023.1303446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/21/2023] [Indexed: 12/29/2023] Open
Abstract
Introduction Pre-erythrocytic malaria vaccines hold the promise of inducing sterile protection thereby preventing the morbidity and mortality associated with Plasmodium infection. The main surface antigen of P. falciparum sporozoites, i.e., the circumsporozoite protein (CSP), has been extensively explored as a target of such vaccines with significant success in recent years. Systematic adjuvant selection, refinements of the immunization regimen, and physical properties of the antigen may all contribute to the potential of increasing the efficacy of CSP-based vaccines. Protection appears to be dependent in large part on CSP antibodies. However due to a knowledge gap related to the exact correlates of immunity, there is a critical need to improve our ability to down select candidates preclinically before entering clinical trials including with controlled human malaria infections (CHMI). Methods We developed a novel multiplex competition assay based on well-characterized monoclonal antibodies (mAbs) that target crucial epitopes across the CSP molecule. This new tool assesses both, quality and epitope-specific concentrations of vaccine-induced antibodies by measuring their equivalency with a panel of well-characterized, CSP-epitope-specific mAbs. Results Applying this method to RTS,S-immune sera from a CHMI trial demonstrated a quantitative epitope-specificity profile of antibody responses that can differentiate between protected vs. nonprotected individuals. Aligning vaccine efficacy with quantitation of the epitope fine specificity results of this equivalency assay reveals the importance of epitope specificity. Discussion The newly developed serological equivalence assay will inform future vaccine design and possibly even adjuvant selection. This methodology can be adapted to other antigens and disease models, when a panel of relevant mAbs exists, and could offer a unique tool for comparing and down-selecting vaccine formulations.
Collapse
Affiliation(s)
- Jessica S. Bolton
- Biologics Research & Development, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, United States
| | - Randall S. MacGill
- Center for Vaccine Innovation and Access, PATH, Washington, DC, United States
| | - Emily Locke
- Center for Vaccine Innovation and Access, PATH, Washington, DC, United States
| | - Jason A. Regules
- Biologics Research & Development, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, United States
| | - Elke S. Bergmann-Leitner
- Biologics Research & Development, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, United States
| |
Collapse
|
11
|
Tan JH, Ding HX, Fong MY, Lau YL. Genetic diversity and in silico analysis of Plasmodium knowlesi Serine Repeat Antigen (SERA) 3 antigen 2 in Malaysia. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023; 114:105490. [PMID: 37595939 DOI: 10.1016/j.meegid.2023.105490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/29/2023] [Accepted: 08/15/2023] [Indexed: 08/20/2023]
Abstract
Plasmodium knowlesi is the leading cause of malaria in Malaysia. Serine Repeat Antigens (SERAs) have an essential role in the parasite life cycle. However, genetic characterization on P. knowlesi SERA3 Ag2 (PkSERA3 Ag2) is lacking. In the present study, nucleotide diversity, natural selection, and haplotypes of PkSERA3 Ag2 in clinical samples from Peninsular Malaysia and Malaysian Borneo were investigated. A total of 50 P. knowlesi clinical samples were collected from Peninsular Malaysia and Malaysian Borneo. The PkSERA3 Ag2 gene was amplified using PCR, and subsequently cloned and sequenced. Genetic diversity, haplotype, natural selection as well as genetic structure and differentiation of PkSERA3 Ag2 were analysed. In addition, in silico analyses were performed to identify repeat motifs, B-cell epitopes, and antigenicity indices of the protein. Analysis of 114 PkSERA3 Ag2 sequences revealed high nucleotide diversity of the gene in Malaysia. A codon-based Z-test indicated that the gene underwent purifying selection. Haplotype and population structure analyses identified two distinct PkSERA3 Ag2 clusters (K = 2, ΔK = 721.14) but no clear genetic distinction between PkSERA3 Ag2 from Peninsular Malaysia and Malaysian Borneo. FST index indicated moderate differentiation of the gene. In silico analyses revealed unique repeat motifs among PkSERA3 Ag2 isolates. Moreover, the amino acid sequence of PkSERA3 Ag2 exhibited potential B-cell epitopes and possessed high antigenicity indices. These findings enhance the understanding of PkSERA3 Ag2 gene as well as its antigenic properties. Further validation is necessary to ascertain the utility of PkSERA3 Ag2 as a serological marker for P. knowlesi infection.
Collapse
Affiliation(s)
- Jia Hui Tan
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Hans Xing Ding
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Mun Yik Fong
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Yee Ling Lau
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
12
|
Watson FN, Shears MJ, Kalata AC, Duncombe CJ, Seilie AM, Chavtur C, Conrad E, Talavera IC, Raappana A, Sather DN, Chakravarty S, Sim BKL, Hoffman SL, Tsuji M, Murphy SC. Ultra-low volume intradermal administration of radiation-attenuated sporozoites with the glycolipid adjuvant 7DW8-5 completely protects mice against malaria. RESEARCH SQUARE 2023:rs.3.rs-3243319. [PMID: 37609210 PMCID: PMC10441511 DOI: 10.21203/rs.3.rs-3243319/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Malaria is caused by Plasmodium parasites and was responsible for over 247 million infections and 619,000 deaths in 2021. Radiation-attenuated sporozoite (RAS) vaccines can completely prevent blood stage infection by inducing protective liver-resident memory CD8+ T cells. Such T cells can be induced by 'prime-and-trap' vaccination, which here combines DNA priming against the P. yoelii circumsporozoite protein (CSP) with a subsequent intravenous (IV) dose of liver-homing RAS to "trap" the activated and expanding T cells in the liver. Prime-and-trap confers durable protection in mice, and efforts are underway to translate this vaccine strategy to the clinic. However, it is unclear whether the RAS trapping dose must be strictly administered by the IV route. Here we show that intradermal (ID) RAS administration can be as effective as IV administration if RAS are co-administrated with the glycolipid adjuvant 7DW8-5 in an ultra-low inoculation volume. In mice, the co-administration of RAS and 7DW8-5 in ultra-low ID volumes (2.5 μL) was completely protective and dose sparing compared to standard volumes (10-50 μL) and induced protective levels of CSP-specific CD8+ T cells in the liver. Our finding that adjuvants and ultra-low volumes are required for ID RAS efficacy may explain why prior reports about higher volumes of unadjuvanted ID RAS proved less effective. The ID route may offer significant translational advantages over the IV route and could improve sporozoite vaccine development.
Collapse
|
13
|
Martin GM, Fernández-Quintero ML, Lee WH, Pholcharee T, Eshun-Wilson L, Liedl KR, Pancera M, Seder RA, Wilson IA, Ward AB. Structural basis of epitope selectivity and potent protection from malaria by PfCSP antibody L9. Nat Commun 2023; 14:2815. [PMID: 37198165 PMCID: PMC10192352 DOI: 10.1038/s41467-023-38509-2] [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: 12/12/2022] [Accepted: 05/04/2023] [Indexed: 05/19/2023] Open
Abstract
A primary objective in malaria vaccine design is the generation of high-quality antibody responses against the circumsporozoite protein of the malaria parasite, Plasmodium falciparum (PfCSP). To enable rational antigen design, we solved a cryo-EM structure of the highly potent anti-PfCSP antibody L9 in complex with recombinant PfCSP. We found that L9 Fab binds multivalently to the minor (NPNV) repeat domain, which is stabilized by a unique set of affinity-matured homotypic, antibody-antibody contacts. Molecular dynamics simulations revealed a critical role of the L9 light chain in integrity of the homotypic interface, which likely impacts PfCSP affinity and protective efficacy. These findings reveal the molecular mechanism of the unique NPNV selectivity of L9 and emphasize the importance of anti-homotypic affinity maturation in protective immunity against P. falciparum.
Collapse
Affiliation(s)
- Gregory M Martin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Monica L Fernández-Quintero
- Department of General, Inorganic, and Theoretical Chemistry, Center for Chemistry and Biomedicine, The University of Innsbruck; Innrain 80-82/III, 6020, Innsbruck, Austria
| | - Wen-Hsin Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Tossapol Pholcharee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Biochemistry, University of Oxford, Oxford, OX1 3DR, UK
| | - Lisa Eshun-Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Klaus R Liedl
- Department of General, Inorganic, and Theoretical Chemistry, Center for Chemistry and Biomedicine, The University of Innsbruck; Innrain 80-82/III, 6020, Innsbruck, Austria
| | - Marie Pancera
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
14
|
Tripathi P, Bender MF, Lei H, Da Silva Pereira L, Shen CH, Bonilla B, Dillon M, Ou L, Pancera M, Wang LT, Zhang B, Batista FD, Idris AH, Seder RA, Kwong PD. Cryo-EM structures of anti-malarial antibody L9 with circumsporozoite protein reveal trimeric L9 association and complete 27-residue epitope. Structure 2023; 31:480-491.e4. [PMID: 36931276 PMCID: PMC10237622 DOI: 10.1016/j.str.2023.02.009] [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: 11/03/2022] [Revised: 01/17/2023] [Accepted: 02/13/2023] [Indexed: 03/18/2023]
Abstract
Monoclonal antibody L9 recognizes the Plasmodium falciparum circumsporozoite protein (PfCSP) and is highly protective following controlled human malaria challenge. To gain insight into its function, we determined cryoelectron microscopy (cryo-EM) structures of L9 in complex with full-length PfCSP and assessed how this recognition influenced protection by wild-type and mutant L9s. Cryo-EM reconstructions at 3.6- and 3.7-Å resolution revealed L9 to recognize PfCSP as an atypical trimer. Each of the three L9s in the trimer directly recognized an Asn-Pro-Asn-Val (NPNV) tetrapeptide on PfCSP and interacted homotypically to facilitate L9-trimer assembly. We analyzed peptides containing different repeat tetrapeptides for binding to wild-type and mutant L9s to delineate epitope and homotypic components of L9 recognition; we found both components necessary for potent malaria protection. Last, we found the 27-residue stretch recognized by L9 to be highly conserved in P. falciparum isolates, suggesting the newly revealed complete L9 epitope to be an attractive vaccine target.
Collapse
Affiliation(s)
- Prabhanshu Tripathi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael F Bender
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Haotian Lei
- Research Technology Branch Electron Microscopy Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lais Da Silva Pereira
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chen-Hsiang Shen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brian Bonilla
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marlon Dillon
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Li Ou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marie Pancera
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Lawrence T Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Baoshan Zhang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Facundo D Batista
- Departments of Immunology and Microbiology, Harvard Medical School, Boston, MA 02139, USA
| | - Azza H Idris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
15
|
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
|
16
|
Ngulube P. Humoral Immune Responses to P. falciparum Circumsporozoite Protein (Pfcsp) Induced by the RTS, S Vaccine - Current Update. Infect Drug Resist 2023; 16:2147-2157. [PMID: 37077252 PMCID: PMC10106824 DOI: 10.2147/idr.s401247] [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: 12/13/2022] [Accepted: 03/23/2023] [Indexed: 04/21/2023] Open
Abstract
Malaria vaccines targeting the circumsporozoite protein (CSP) of the P. falciparum parasite have been overall relatively promising. RTS, S is a pre-erythrocytic recombinant protein-based malaria vaccine that targets CSP. RTS, S effectiveness shows some limited success regardless of its 58% efficacy for severe disease. P. falciparum circumsporozoite protein (Pfcsp) has stood to be the main candidate protein for most pre-erythrocytic stage vaccines. Studies on the structural and biophysical characteristics of antibodies specific to CSP (anti-CSP) are underway to achieve fine specificity with the CSP polymorphic regions. More recent studies have proposed the use of different kinds of monoclonal antibodies, the use of appropriate adjuvants, ideal vaccination dose and frequency, and improved targeting of particular epitopes for the robust production of functional antibodies and high complement-fixing activity as other potential methods for achieving long-lasting RTS, S. This review highlights recent findings regarding humoral immune responses to CSP elicited by RTS, S vaccine.
Collapse
Affiliation(s)
- Peter Ngulube
- Department of Biological Sciences, Academy of Medical Sciences, Malawi University of Science and Technology, Thyolo, Malawi
- Correspondence: Peter Ngulube, Email
| |
Collapse
|
17
|
de Korne CM, van Schuijlenburg R, Sijtsma JC, de Bes HM, Baalbergen E, Azargoshasb S, van Oosterom MN, McCall MBB, van Leeuwen FWB, Roestenberg M. Sporozoite motility as a quantitative readout for anti-CSP antibody inhibition. Sci Rep 2022; 12:17194. [PMID: 36229488 PMCID: PMC9561690 DOI: 10.1038/s41598-022-22154-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/10/2022] [Indexed: 01/05/2023] Open
Abstract
Antibodies can prevent malaria by neutralizing the infectious Plasmodium falciparum sporozoites (SPZ) before they establish an infection in the liver. Circumsporozoite protein (CSP), the most abundant surface protein of SPZ is the leading candidate for passive (and subunit) immunization approaches against malaria. Comprehensive assessment of the parasite-inhibitory capacity of anti-CSP monoclonal antibodies (mAbs) is an important step in advancing CSP-based immunization strategies. In this study, we employed a quantitative imaging-based motility assay to quantify the effect of anti-CSP mAbs on SPZ motility, both in vitro and in human skin.Our assay provided a quantitative measure of mAb parasite-inhibitory capacity through measurement of the half-maximal motility inhibitory concentration (IC50M) value for anti-CSP mAbs (IC50M 2A10: 24 nM, IC50M 3SP2: 71 nM). We found a sevenfold discrepancy between the IC50M and the binding saturation concentration measured by ELISA, possibly related to the observed shedding of CSP-mAb complexes during SPZ movement. In a subset of SPZ (5%), in vitro motility was unaffected by the presence of 2A10 while 3SP2 was able to completely block movement. In our ex vivo skin explant model, SPZ proved less susceptible to anti-CSP mAbs compared to SPZ in an in vitro environment. By quantitatively assessing motility, we created a valuable tool that can be used for comprehensive assessment of anti-CSP mAb potency. Insight that will help deepen our understanding of anti-CSP mAb potency and guide selection of the most promising anti-CSP mAbs for downstream clinical development.
Collapse
Affiliation(s)
- C M de Korne
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - R van Schuijlenburg
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - J C Sijtsma
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - H M de Bes
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - E Baalbergen
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - S Azargoshasb
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - M N van Oosterom
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - M B B McCall
- Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - F W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - M Roestenberg
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
18
|
He ZQ, Zhang QQ, Wang D, Hu YB, Zhou RM, Qian D, Yang CY, Lu DL, Li SH, Liu Y, Zhang HW. Genetic polymorphism of circumsporozoite protein of Plasmodium falciparum among Chinese migrant workers returning from Africa to Henan Province. Malar J 2022; 21:248. [PMID: 36030242 PMCID: PMC9419638 DOI: 10.1186/s12936-022-04275-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 08/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Plasmodium falciparum malaria is recognized as a major global public health problem. The malaria vaccine was important because the case fatality rate of falciparum malaria was high. Plasmodium falciparum circumsporozoite protein (PfCSP) is one of the potential vaccine candidates, but the genetic polymorphism of PfCSP raises concerns regarding the efficacy of the vaccine. This study aimed to investigate the genetic polymorphism of PfCSP and provide data for the improvement of PfCSP-based vaccine (RTS,S malaria vaccine). METHODS Blood samples were collected from 287 Chinese migrant workers who were infected with P. falciparum and returning from Africa to Henan Province during 2016-2018. The Pfcsp genes were analysed to estimate the genetic diversity of this parasite. RESULTS The results showed that there were two mutations at the N-terminus of imported Pfcsp in Henan Province, including insertion amino acids (58.71%, 118/201) and A → G (38.81%, 78/201). The number of repeats of tetrapeptide motifs (NANP/NVDP/NPNP/NVDA) in the central repeat region ranged mainly from 39 to 42 (97.51%, 196/201). A total of 14 nonsynonymous amino acid changes were found at the C-terminus. The average nucleotide difference (K) of imported Pfcsp in Henan Province was 5.719, and the haplotype diversity (Hd) was 0.964 ± 0.004. The estimated value of dN-dS was 0.047, indicating that the region may be affected by positive natural selection. The minimum number of recombination events (Rm) of imported Pfcsp in Henan Province was close to that in Africa. The analysis of genetic differentiation showed that there may be moderate differentiation between East Africa and North Africa (Fst = 0.06484), and the levels of differentiation in the other regions were very small (Fst < 0.05). CONCLUSIONS The N-terminus of Pfcsp was relatively conserved, and the central repeat region and the Th2R and Th3R regions of the C-terminus were highly polymorphic. The gene polymorphism pattern among Chinese migrant workers returning from Africa to Henan Province was consistent with that in Africa. The geographical pattern of population differentiation and the evidence of natural selection and gene recombination suggested that the effect of polymorphism on the efficacy of PfCSP-based vaccines should be considered.
Collapse
Affiliation(s)
- Zhi-Quan He
- Department of Parasite Disease Control and Prevention, Henan Center for Disease Control and Prevention, Zhengzhou, China.,Henan Key Laboratory of Pathogenic Microorganisms, No. 105 South Agricultural Road, Zhengdong New District, Zhengzhou, 450016, China
| | - Qun-Qun Zhang
- Fengtai District Center for Disease Control and Prevention, Beijing, China
| | - Dan Wang
- Department of Parasite Disease Control and Prevention, Henan Center for Disease Control and Prevention, Zhengzhou, China.,Henan Key Laboratory of Pathogenic Microorganisms, No. 105 South Agricultural Road, Zhengdong New District, Zhengzhou, 450016, China
| | - Ya-Bo Hu
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Rui-Min Zhou
- Department of Parasite Disease Control and Prevention, Henan Center for Disease Control and Prevention, Zhengzhou, China.,Henan Key Laboratory of Pathogenic Microorganisms, No. 105 South Agricultural Road, Zhengdong New District, Zhengzhou, 450016, China
| | - Dan Qian
- Department of Parasite Disease Control and Prevention, Henan Center for Disease Control and Prevention, Zhengzhou, China.,Henan Key Laboratory of Pathogenic Microorganisms, No. 105 South Agricultural Road, Zhengdong New District, Zhengzhou, 450016, China
| | - Cheng-Yun Yang
- Department of Parasite Disease Control and Prevention, Henan Center for Disease Control and Prevention, Zhengzhou, China.,Henan Key Laboratory of Pathogenic Microorganisms, No. 105 South Agricultural Road, Zhengdong New District, Zhengzhou, 450016, China
| | - De-Ling Lu
- Department of Parasite Disease Control and Prevention, Henan Center for Disease Control and Prevention, Zhengzhou, China.,Henan Key Laboratory of Pathogenic Microorganisms, No. 105 South Agricultural Road, Zhengdong New District, Zhengzhou, 450016, China
| | - Su-Hua Li
- Department of Parasite Disease Control and Prevention, Henan Center for Disease Control and Prevention, Zhengzhou, China.,Henan Key Laboratory of Pathogenic Microorganisms, No. 105 South Agricultural Road, Zhengdong New District, Zhengzhou, 450016, China
| | - Ying Liu
- Department of Parasite Disease Control and Prevention, Henan Center for Disease Control and Prevention, Zhengzhou, China. .,Henan Key Laboratory of Pathogenic Microorganisms, No. 105 South Agricultural Road, Zhengdong New District, Zhengzhou, 450016, China.
| | - Hong-Wei Zhang
- Department of Parasite Disease Control and Prevention, Henan Center for Disease Control and Prevention, Zhengzhou, China. .,Henan Key Laboratory of Pathogenic Microorganisms, No. 105 South Agricultural Road, Zhengdong New District, Zhengzhou, 450016, China.
| |
Collapse
|
19
|
Tursi NJ, Reeder SM, Flores-Garcia Y, Bah MA, Mathis-Torres S, Salgado-Jimenez B, Esquivel R, Xu Z, Chu JD, Humeau L, Patel A, Zavala F, Weiner DB. Engineered DNA-encoded monoclonal antibodies targeting Plasmodium falciparum circumsporozoite protein confer single dose protection in a murine malaria challenge model. Sci Rep 2022; 12:14313. [PMID: 35995959 PMCID: PMC9395511 DOI: 10.1038/s41598-022-18375-6] [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: 03/01/2022] [Accepted: 05/06/2022] [Indexed: 12/15/2022] Open
Abstract
Novel approaches for malaria prophylaxis remain important. Synthetic DNA-encoded monoclonal antibodies (DMAbs) are a promising approach to generate rapid, direct in vivo host-generated mAbs with potential benefits in production simplicity and distribution coupled with genetic engineering. Here, we explore this approach in a malaria challenge model. We engineered germline-reverted DMAbs based on human mAb clones CIS43, 317, and L9 which target a junctional epitope, major repeat, and minor repeat of the Plasmodium falciparum circumsporozoite protein (CSP) respectively. DMAb variants were encoded into a plasmid vector backbone and their expression and binding profiles were characterized. We demonstrate long-term serological expression of DMAb constructs resulting in in vivo efficacy of CIS43 GL and 317 GL in a rigorous mosquito bite mouse challenge model. Additionally, we engineered an Fc modified variant of CIS43 and L9-based DMAbs to ablate binding to C1q to test the impact of complement-dependent Fc function on challenge outcomes. Complement knockout variant DMAbs demonstrated similar protection to that of WT Fc DMAbs supporting the notion that direct binding to the parasite is sufficient for the protection observed. Further investigation of DMAbs for malaria prophylaxis appears of importance.
Collapse
Affiliation(s)
- Nicholas J Tursi
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sophia M Reeder
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Mamadou A Bah
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Shamika Mathis-Torres
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Berenice Salgado-Jimenez
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Rianne Esquivel
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Ziyang Xu
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jacqueline D Chu
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Laurent Humeau
- Inovio Pharmaceuticals, Plymouth Meeting, PA, 19462, USA
| | - Ami Patel
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - David B Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA.
| |
Collapse
|
20
|
Vaccine co-display of CSP and Pfs230 on liposomes targeting two Plasmodium falciparum differentiation stages. Commun Biol 2022; 5:773. [PMID: 35915227 PMCID: PMC9341416 DOI: 10.1038/s42003-022-03688-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 07/08/2022] [Indexed: 12/13/2022] Open
Abstract
A vaccine targeting multiple stages of the Plasmodium falciparum parasite life cycle is desirable. The sporozoite surface Circumsporozoite Protein (CSP) is the target of leading anti-infective P. falciparum pre-erythrocytic vaccines. Pfs230, a sexual-stage P. falciparum surface protein, is currently in trials as the basis for a transmission-blocking vaccine, which inhibits parasite development in the mosquito vector. Here, recombinant full-length CSP and a Pfs230 fragment (Pfs230D1+) are co-displayed on immunogenic liposomes to induce immunity against both infection and transmission. Liposomes contain cobalt-porphyrin phospholipid (CoPoP), monophosphoryl lipid A and QS-21, and rapidly bind His-tagged CSP and Pfs230D1+ upon admixture to form bivalent particles that maintain reactivity with conformational monoclonal antibodies. Use of multicolor fluorophore-labeled antigens reveals liposome binding upon admixture, stability in serum and enhanced uptake in murine macrophages in vitro. Bivalent liposomes induce humoral and cellular responses against both CSP and Pfs230D1+. Vaccine-induced antibodies reduce parasite numbers in mosquito midguts in a standard membrane feeding assay. Mice immunized with liposome-displayed antigens or that passively receive antibodies from immunized rabbits have reduced parasite liver burden following challenge with transgenic sporozoites expressing P. falciparum CSP.
Collapse
|
21
|
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
|
22
|
Arias CF, Acosta FJ, Fernandez-Arias C. Killing the competition: a theoretical framework for liver-stage malaria. Open Biol 2022; 12:210341. [PMID: 35350863 PMCID: PMC8965401 DOI: 10.1098/rsob.210341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The first stage of malaria infections takes place inside the host's hepatocytes. Remarkably, Plasmodium parasites do not infect hepatocytes immediately after reaching the liver. Instead, they migrate through several hepatocytes before infecting their definitive host cells, thus increasing their chances of immune destruction. Considering that malaria can proceed normally without cell traversal, this is indeed a puzzling behaviour. In fact, the role of hepatocyte traversal remains unknown to date, implying that the current understanding of malaria is incomplete. In this work, we hypothesize that the parasites traverse hepatocytes to actively trigger an immune response in the host. This behaviour would be part of a strategy of superinfection exclusion aimed to reduce intraspecific competition during the blood stage of the infection. Based on this hypothesis, we formulate a comprehensive theory of liver-stage malaria that integrates all the available knowledge about the infection. The interest of this new paradigm is not merely theoretical. It highlights major issues in the current empirical approach to the study of Plasmodium and suggests new strategies to fight malaria.
Collapse
Affiliation(s)
- Clemente F. Arias
- Centro de Investigaciones Biológicas (CSIC), Madrid, Spain,Grupo Interdisciplinar de Sistemas Complejos de Madrid, Spain
| | | | - Cristina Fernandez-Arias
- Departamento de Inmunología, Universidad Complutense de Madrid, Spain,Instituto de Medicina Molecular, Universidade de Lisboa, Portugal
| |
Collapse
|
23
|
Wang LT, Hurlburt NK, Schön A, Flynn BJ, Flores-Garcia Y, Pereira LS, Kiyuka PK, Dillon M, Bonilla B, Zavala F, Idris AH, Francica JR, Pancera M, Seder RA. The light chain of the L9 antibody is critical for binding circumsporozoite protein minor repeats and preventing malaria. Cell Rep 2022; 38:110367. [PMID: 35172158 PMCID: PMC8896312 DOI: 10.1016/j.celrep.2022.110367] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/17/2021] [Accepted: 01/20/2022] [Indexed: 01/23/2023] Open
Abstract
L9 is a potent human monoclonal antibody (mAb) that preferentially binds two adjacent NVDP minor repeats and cross-reacts with NANP major repeats of the Plasmodium falciparum circumsporozoite protein (PfCSP) on malaria-infective sporozoites. Understanding this mAb's ontogeny and mechanisms of binding PfCSP will facilitate vaccine development. Here, we isolate mAbs clonally related to L9 and show that this B cell lineage has baseline NVDP affinity and evolves to acquire NANP reactivity. Pairing the L9 kappa light chain (L9κ) with clonally related heavy chains results in chimeric mAbs that cross-link two NVDPs, cross-react with NANP, and more potently neutralize sporozoites in vivo compared with their original light chain. Structural analyses reveal that the chimeric mAbs bound minor repeats in a type-1 β-turn seen in other repeat-specific antibodies. These data highlight the importance of L9κ in binding NVDP on PfCSP to neutralize sporozoites and suggest that PfCSP-based immunogens might be improved by presenting ≥2 NVDPs.
Collapse
Affiliation(s)
- Lawrence T Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicholas K Hurlburt
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Arne Schön
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Barbara J Flynn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Lais S Pereira
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Patience K Kiyuka
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marlon Dillon
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brian Bonilla
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Azza H Idris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joseph R Francica
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marie Pancera
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
24
|
|