1
|
Naghizadeh M, Miura K, Addo Ofori E, Long C, Sagara I, Tiono AB, Plieskatt J, Theisen M. Magnitude and durability of ProC6C-AlOH/Matrix-M tm vaccine-induced malaria transmission-blocking antibodies in Burkinabe adults from a Phase 1 randomized trial. Hum Vaccin Immunother 2025; 21:2488075. [PMID: 40208198 PMCID: PMC11988263 DOI: 10.1080/21645515.2025.2488075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/03/2025] [Accepted: 03/30/2025] [Indexed: 04/11/2025] Open
Abstract
ProC6C is a multi-stage malaria vaccine designed to disrupt parasite transmission and prevent infection by incorporating three parasite proteins (Pfs230-Pro, Pfs48/45-6C, and CSP) in a single vaccine antigen. The Phase 1 clinical trial (PACTR202201848463189) conducted in Burkina Faso, showed ProC6C-AlOH/Matrix-M was safe, well tolerated, immunogenic and generated a functional antibody response to all three constituent antigens at the primary output (D70). As magnitude and durability are central to an efficacious malaria vaccine, analysis was expanded past the initial endpoint, to determine transmission-blocking antibodies (anti-Pfs230 and anti-Pfs48/45-6C) present through D180. Analysis of transmission-reducing activity (TRA) showed 7/20 samples remained biologically active at D180. To identify immune biomarkers for high levels of TRA, the Pfs48/45-6C IgG concentration (calculated relative to the transmission-blocking mAb TB31F) was compared among TRA positive and negative individuals. The magnitude of anti-Pfs48/45-6C IgG had an excellent predictive accuracy (area under the receiver operating curve [ROC AUC] >0.8) with a threshold of 8.7 μg/ml for significant TRA. Additionally, there was significant correlation of TRA and anti-Pfs48/45 epitope I IgG concentration but not significant correlation for anti-Pfs230-Pro IgG, suggesting that vaccine-induced anti-Pfs48/45-6C IgG is the main predictor of TRA. This finding was corroborated by the observation that complement had no effect on TRA in the standard membrane feeding assay (SMFA). Collectively, these efforts confirm the transmission-blocking attributes of ProC6C and suggest that an alternative dosing regimen be evaluated in future clinical trials to improve longevity of functional transmission-reducing antibodies.
Collapse
Affiliation(s)
- 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, USA
| | - 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
| | - Carole Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - 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
| | - Alfred B. Tiono
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Jordan Plieskatt
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
| | - 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
|
2
|
Kucharska I, Ivanochko D, Hailemariam S, Inklaar MR, Kim HR, Teelen K, Stoter R, van de Vegte-Bolmer M, van Gemert GJ, Semesi A, McLeod B, Ki A, Lee WK, Rubinstein JL, Jore MM, Julien JP. Structural elucidation of full-length Pfs48/45 in complex with potent monoclonal antibodies isolated from a naturally exposed individual. Nat Struct Mol Biol 2025:10.1038/s41594-025-01532-6. [PMID: 40404982 DOI: 10.1038/s41594-025-01532-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/12/2025] [Indexed: 05/24/2025]
Abstract
Biomedical interventions that block the transmission of Plasmodium falciparum (Pf) from humans to mosquitoes may be critical for malaria elimination. Pfs48/45, a gamete-surface protein essential for Pf development in the mosquito midgut, is a target of clinical-stage transmission-blocking vaccines and monoclonal antibodies (mAbs) that disrupt Pf transmission to mosquitoes. Antibodies directed to domain 3 of Pfs48/45 have been structurally and functionally described; however, in-depth information about other inhibitory epitopes on Pfs48/45 is currently limited. Here, we present a cryo-electron microscopy structure of full-length Pfs48/45 in complex with potent human mAbs targeting all three domains. Our data indicate that although Pfs48/45 domains 1 and 2 are rigidly coupled, there is substantial conformational flexibility between domains 2 and 3. Characterization of mAbs against domain 1 revealed the presence of a conformational epitope class that is largely conserved across Pf field isolates and is associated with recognition by potent antibodies. Our study provides insights into epitopes across full-length Pfs48/45 and has implications for the design of next-generation malaria interventions.
Collapse
Affiliation(s)
- Iga Kucharska
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Danton Ivanochko
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Sophia Hailemariam
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Maartje R Inklaar
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hee Ryung Kim
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Karina Teelen
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rianne Stoter
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anthony Semesi
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Brandon McLeod
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Ahyoung Ki
- Structural Analysis Team, New Drug Development Center, KBIO Osong Medical Innovation Foundation, Osong, Republic of Korea
| | - Won-Kyu Lee
- Structural Analysis Team, New Drug Development Center, KBIO Osong Medical Innovation Foundation, Osong, Republic of Korea
| | - John L Rubinstein
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Matthijs M Jore
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada.
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
3
|
Yoo R, Jore MM, Julien J. Targeting Bottlenecks in Malaria Transmission: Antibody-Epitope Descriptions Guide the Design of Next-Generation Biomedical Interventions. Immunol Rev 2025; 330:e70001. [PMID: 39907429 PMCID: PMC11796336 DOI: 10.1111/imr.70001] [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: 12/26/2024] [Accepted: 01/08/2025] [Indexed: 02/06/2025]
Abstract
Malaria continues to pose a significant burden to global health. Thus, a strong need exists for the development of a diverse panel of intervention strategies and modalities to combat malaria and achieve elimination and eradication goals. Deploying interventions that target bottlenecks in the transmission life cycle of the causative agent of malaria, Plasmodium parasites, is an attractive strategy. The development of highly potent antibody-based biologics, including vaccines, can be greatly facilitated by an in-depth molecular understanding of antibody-epitope interactions. Here, we provide an overview of structurally characterized antibodies targeting lead vaccine candidates expressed during the bottlenecks of the Plasmodium life cycle which include the pre-erythrocytic and sexual stages. The repeat region of the circumsporozoite protein (CSP), domain 1 of Pfs230 and domains 1 and 3 of Pfs48/45 are critical Plasmodium regions targeted by the most potent antibodies at the two bottlenecks of transmission, with other promising targets emerging and requiring further characterization.
Collapse
Affiliation(s)
- Randy Yoo
- Program in Molecular MedicineThe Hospital for Sick Children Research InstituteTorontoOntarioCanada
- Department of BiochemistryUniversity of TorontoTorontoOntarioCanada
| | - Matthijs M. Jore
- Department of Medical MicrobiologyRadboudumcNijmegenThe Netherlands
| | - Jean‐Philippe Julien
- Program in Molecular MedicineThe Hospital for Sick Children Research InstituteTorontoOntarioCanada
- Department of BiochemistryUniversity of TorontoTorontoOntarioCanada
- Department of ImmunologyUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
4
|
Onyango SA, Machani MG, Ochwedo KO, Oriango RM, Lee MC, Kokwaro E, Afrane YA, Githeko AK, Zhong D, Yan G. Plasmodium falciparum Pfs47 haplotype compatibility to Anopheles gambiae in Kisumu, a malaria-endemic region of Kenya. Sci Rep 2025; 15:6550. [PMID: 39994226 PMCID: PMC11850800 DOI: 10.1038/s41598-024-84847-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/27/2024] [Indexed: 02/26/2025] Open
Abstract
Insecticide resistance and outdoor transmission have reduced the effectiveness of existing malaria transmission prevention strategies. As a result, targeted approaches to support continuing malaria control, such as transmission-blocking vaccines, are required. Cross-sectional mass blood screening in children between 5 and 15 years was conducted in Chulaimbo, Kisumu, during the dry and wet seasons in 2018 and 2019. Plasmodium falciparum gametocyte carriers were identified by Microscopy. Subsequently, carriers were used to feed colony bred Anopheles gambiae females in serum replacement and whole blood membrane feeding experiments. The infection prevalence was 19.7% (95% Cl 0.003-0.007) with 95% of the infections being caused by P. falciparum. Of all confirmed P. falciparum infections, 16.9% were gametocytes. Thirty-seven paired experiments showed infection rates of 0.9% and 0.5% in the serum replacement and whole blood experiments, respectively, with no significant difference (P = 0.738). Six Pfs47 haplotypes were identified from 24 sequenced infectious blood samples: Hap_1 (E27D and L240I), Hap_2 (S98T); Hap_3 (E27D); Hap_4 (L240I); Hap_5 (E188D); and Hap_6 without mutations. Haplotype 4 had the highest frequency of 29.2% followed by Hap_3 and Hap_6 at 20.8% each then Hap_1 with a frequency of 16.7%, whereas Hap_5 and Hap_2 had frequencies of 8.3% and 4.2% respectively. Varying frequencies of Pfs47 haplotypes observed from genetically heterogeneous parasite populations in endemic regions illuminates vector compatibility to refracting P. falciparum using the hypothesized lock and key analogy. This acts as a bottleneck that increases the frequency of P. falciparum haplotypes that escape elimination by vector immune responses. The interaction can be used as a potential target for transmission blocking through a refractory host.
Collapse
Affiliation(s)
- Shirley A Onyango
- School of Zoological Sciences, Kenyatta University, Nairobi, Kenya.
- International Centre of Excellence for Malaria Research, Tom Mboya University, Homa Bay, Kenya.
| | - Maxwell G Machani
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Kevin O Ochwedo
- International Centre of Excellence for Malaria Research, Tom Mboya University, Homa Bay, Kenya
- School of Biological Sciences, University of Nairobi, Nairobi, Kenya
| | - Robin M Oriango
- International Centre of Excellence for Malaria Research, Tom Mboya University, Homa Bay, Kenya
| | - Ming-Chieh Lee
- Program in Public Health, College of Health Sciences, University of California at Irvine, Irvine, CA, 92697, USA
| | | | - Yaw A Afrane
- Department of Medical Microbiology, Medical School, University of Ghana, Accra, Ghana
| | - Andrew K Githeko
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Daibin Zhong
- Program in Public Health, College of Health Sciences, University of California at Irvine, Irvine, CA, 92697, USA.
| | - Guiyun Yan
- Program in Public Health, College of Health Sciences, University of California at Irvine, Irvine, CA, 92697, USA
| |
Collapse
|
5
|
Bekkering ET, Yoo R, Hailemariam S, Heide F, Ivanochko D, Jackman M, Proellochs NI, Stoter R, Wanders OT, van Daalen RC, Inklaar MR, Andrade CM, Jansen PW, Vermeulen M, Bousema T, Rubinstein JL, Kooij TW, Jore MM, Julien JP. Structure of endogenous Pfs230:Pfs48/45 in complex with potent malaria transmission-blocking antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638310. [PMID: 39990443 PMCID: PMC11844449 DOI: 10.1101/2025.02.14.638310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The Pfs230:Pfs48/45 complex is essential for malaria parasites to infect mosquitoes and forms the basis for current leading transmission-blocking vaccine candidates, yet little is known about its molecular assembly. Here, we used cryogenic electron microscopy to elucidate the structure of the endogenous Pfs230:Pfs48/45 complex bound to six potent transmission-blocking antibodies. Pfs230 consists of multiple domain clusters rigidified by interactions mediated through insertion domains. Membrane-anchored Pfs48/45 forms a disc-like structure and interacts with a short C-terminal peptide on Pfs230 that is critical for Pfs230 membrane-retention in vivo. Analyses of Pfs48/45- and Pfs230-targeted antibodies identify conserved epitopes on the Pfs230:Pfs48/45 complex and provides a structural paradigm for complement-dependent activity of Pfs230-targeting antibodies. Altogether, the Pfs230:Pfs48/45 antibody-complex structure presented improves our understanding of malaria transmission biology and the mechanisms of action of transmission-blocking antibodies, informing the development of next-generation transmission-blocking interventions.
Collapse
Affiliation(s)
- Ezra T. Bekkering
- Department of Medical Microbiology, Radboud University Medical Center, The Netherlands
| | - Randy Yoo
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Sophia Hailemariam
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Fabian Heide
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Danton Ivanochko
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Matthew Jackman
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | | | - Rianne Stoter
- Department of Medical Microbiology, Radboud University Medical Center, The Netherlands
| | - Oscar T. Wanders
- Department of Medical Microbiology, Radboud University Medical Center, The Netherlands
| | - Renate C. van Daalen
- Department of Medical Microbiology, Radboud University Medical Center, The Netherlands
| | - Maartje R. Inklaar
- Department of Medical Microbiology, Radboud University Medical Center, The Netherlands
| | - Carolina M. Andrade
- Department of Medical Microbiology, Radboud University Medical Center, The Netherlands
| | - Pascal W.T.C. Jansen
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Teun Bousema
- Department of Medical Microbiology, Radboud University Medical Center, The Netherlands
| | - John L. Rubinstein
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Taco W.A. Kooij
- Department of Medical Microbiology, Radboud University Medical Center, The Netherlands
| | - Matthijs M. Jore
- Department of Medical Microbiology, Radboud University Medical Center, The Netherlands
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Amen A, Yoo R, Fabra-García A, Bolscher J, Stone WJR, Bally I, Dergan-Dylon S, Kucharska I, de Jong RM, de Bruijni M, Bousema T, King CR, MacGill RS, Sauerwein RW, Julien JP, Poignard P, Jore MM. Target-agnostic identification of human antibodies to Plasmodium falciparum sexual forms reveals cross-stage recognition of glutamate-rich repeats. eLife 2025; 13:RP97865. [PMID: 39817720 PMCID: PMC11737873 DOI: 10.7554/elife.97865] [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: 01/18/2025] Open
Abstract
Circulating sexual stages of Plasmodium falciparum (Pf) can be transmitted from humans to mosquitoes, thereby furthering the spread of malaria in the population. It is well established that antibodies can efficiently block parasite transmission. In search for naturally acquired antibodies targets on sexual stages, we established an efficient method for target-agnostic single B cell activation followed by high-throughput selection of human monoclonal antibodies (mAbs) reactive to sexual stages of Pf in the form of gametes and gametocyte extracts. We isolated mAbs reactive against a range of Pf proteins including well-established targets Pfs48/45 and Pfs230. One mAb, B1E11K, was cross-reactive to various proteins containing glutamate-rich repetitive elements expressed at different stages of the parasite life cycle. A crystal structure of two B1E11K Fab domains in complex with its main antigen, RESA, expressed on asexual blood stages, showed binding of B1E11K to a repeating epitope motif in a head-to-head conformation engaging in affinity-matured homotypic interactions. Thus, this mode of recognition of Pf proteins, previously described only for Pf circumsporozoite protein (PfCSP), extends to other repeats expressed across various stages. The findings augment our understanding of immune-pathogen interactions to repeating elements of the Plasmodium parasite proteome and underscore the potential of the novel mAb identification method used to provide new insights into the natural humoral immune response against Pf.
Collapse
Affiliation(s)
- Axelle Amen
- CNRS, Université Grenoble Alpes, CEA, UMR5075, Institut de Biologie StructuraleGrenobleFrance
- CHU Grenoble AlpesGrenobleFrance
| | - Randy Yoo
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
- Department of Biochemistry, University of TorontoTorontoCanada
| | - Amanda Fabra-García
- Department of Medical Microbiology, Radboud University Medical CenterNijmegenNetherlands
| | | | - William JR Stone
- Department of Immunology and Infection, London School of Hygiene and Tropical MedicineLondonUnited Kingdom
| | - Isabelle Bally
- CNRS, Université Grenoble Alpes, CEA, UMR5075, Institut de Biologie StructuraleGrenobleFrance
| | - Sebastián Dergan-Dylon
- CNRS, Université Grenoble Alpes, CEA, UMR5075, Institut de Biologie StructuraleGrenobleFrance
| | - Iga Kucharska
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
| | - Roos M de Jong
- Department of Medical Microbiology, Radboud University Medical CenterNijmegenNetherlands
| | | | - Teun Bousema
- Department of Medical Microbiology, Radboud University Medical CenterNijmegenNetherlands
| | - C Richter King
- Center for Vaccine Innovation and Access, PATHWashington D.C.United States
| | - Randall S MacGill
- Center for Vaccine Innovation and Access, PATHWashington D.C.United States
| | | | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
- Department of Biochemistry, University of TorontoTorontoCanada
- Department of Immunology, University of TorontoTorontoCanada
| | - Pascal Poignard
- CNRS, Université Grenoble Alpes, CEA, UMR5075, Institut de Biologie StructuraleGrenobleFrance
- CHU Grenoble AlpesGrenobleFrance
| | - Matthijs M Jore
- Department of Medical Microbiology, Radboud University Medical CenterNijmegenNetherlands
| |
Collapse
|
7
|
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
|
8
|
Tang WK, Salinas ND, Kolli SK, Xu S, Urusova DV, Kumar H, Jimah JR, Subramani PA, Ogbondah MM, Barnes SJ, Adams JH, Tolia NH. Multistage protective anti-CelTOS monoclonal antibodies with cross-species sterile protection against malaria. Nat Commun 2024; 15:7487. [PMID: 39209843 PMCID: PMC11362571 DOI: 10.1038/s41467-024-51701-2] [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: 02/05/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
CelTOS is a malaria vaccine antigen that is conserved in Plasmodium and other apicomplexan parasites and plays a role in cell-traversal. The structural basis and mechanisms of CelTOS-induced protective immunity to parasites are unknown. Here, CelTOS-specific monoclonal antibodies (mAbs) 7g7 and 4h12 demonstrated multistage activity, protecting against liver infection and preventing parasite transmission to mosquitoes. Both mAbs demonstrated cross-species activity with sterile protection against in vivo challenge with transgenic parasites containing either P. falciparum or P. vivax CelTOS, and with transmission reducing activity against P. falciparum. The mAbs prevented CelTOS-mediated pore formation providing insight into the protective mechanisms. X-ray crystallography and mutant-library epitope mapping revealed two distinct broadly conserved neutralizing epitopes. 7g7 bound to a parallel dimer of CelTOS, while 4h12 bound to a novel antiparallel dimer architecture. These findings inform the design of antibody therapies and vaccines and raise the prospect of a single intervention to simultaneously combat P. falciparum and P. vivax malaria.
Collapse
MESH Headings
- Antibodies, Monoclonal/immunology
- Animals
- Plasmodium falciparum/immunology
- Plasmodium vivax/immunology
- Malaria Vaccines/immunology
- Antibodies, Protozoan/immunology
- Mice
- Malaria, Falciparum/immunology
- Malaria, Falciparum/prevention & control
- Malaria, Falciparum/parasitology
- Crystallography, X-Ray
- Epitopes/immunology
- Malaria, Vivax/prevention & control
- Malaria, Vivax/immunology
- Malaria, Vivax/parasitology
- Antigens, Protozoan/immunology
- Humans
- Female
- Epitope Mapping
- Malaria/immunology
- Malaria/prevention & control
- Malaria/parasitology
- Mice, Inbred BALB C
- Protozoan Proteins/immunology
- Protozoan Proteins/chemistry
Collapse
Affiliation(s)
- Wai Kwan Tang
- Host‒Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nichole D Salinas
- Host‒Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Surendra Kumar Kolli
- Center of Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Shulin Xu
- Center of Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Darya V Urusova
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hirdesh Kumar
- Host‒Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John R Jimah
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Pradeep Annamalai Subramani
- Center of Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Madison M Ogbondah
- Center of Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Samantha J Barnes
- Center of Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, FL, USA
| | - John H Adams
- Center of Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Niraj H Tolia
- Host‒Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
9
|
Ciubotariu II, Broyles BK, Xie S, Thimmapuram J, Mwenda MC, Mambwe B, Mulube C, Matoba J, Schue JL, Moss WJ, Bridges DJ, He Q, Carpi G. Diversity and selection analyses identify transmission-blocking antigens as the optimal vaccine candidates in Plasmodium falciparum. EBioMedicine 2024; 106:105227. [PMID: 39018754 PMCID: PMC11663769 DOI: 10.1016/j.ebiom.2024.105227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND A highly effective vaccine for malaria remains an elusive target, at least in part due to the under-appreciated natural parasite variation. This study aimed to investigate genetic and structural variation, and immune selection of leading malaria vaccine candidates across the Plasmodium falciparum's life cycle. METHODS We analysed 325 P. falciparum whole genome sequences from Zambia, in addition to 791 genomes from five other African countries available in the MalariaGEN Pf3k Database. Ten vaccine antigens spanning three life-history stages were examined for genetic and structural variations, using population genetics measures, haplotype network analysis, and 3D structure selection analysis. FINDINGS Among the ten antigens analysed, only three in the transmission-blocking vaccine category display P. falciparum 3D7 as the dominant haplotype. The antigens AMA1, CSP, MSP119 and CelTOS, are much more diverse than the other antigens, and their epitope regions are under moderate to strong balancing selection. In contrast, Rh5, a blood stage antigen, displays low diversity yet slightly stronger immune selection in the merozoite-blocking epitope region. Except for CelTOS, the transmission-blocking antigens Pfs25, Pfs48/45, Pfs230, Pfs47, and Pfs28 exhibit minimal diversity and no immune selection in epitopes that induce strain-transcending antibodies, suggesting potential effectiveness of 3D7-based vaccines in blocking transmission. INTERPRETATION These findings offer valuable insights into the selection of optimal vaccine candidates against P. falciparum. Based on our results, we recommend prioritising conserved merozoite antigens and transmission-blocking antigens. Combining these antigens in multi-stage approaches may be particularly promising for malaria vaccine development initiatives. FUNDING Purdue Department of Biological Sciences; Puskas Memorial Fellowship; National Institute of Allergy and Infectious Diseases (U19AI089680).
Collapse
Affiliation(s)
- Ilinca I Ciubotariu
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Bradley K Broyles
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Shaojun Xie
- Bioinformatics Core, Purdue University, West Lafayette, IN, USA
| | | | - Mulenga C Mwenda
- PATH-Malaria Control and Elimination Partnership in Africa (MACEPA), National Malaria Elimination Centre, Lusaka, Zambia
| | - Brenda Mambwe
- PATH-Malaria Control and Elimination Partnership in Africa (MACEPA), National Malaria Elimination Centre, Lusaka, Zambia
| | - Conceptor Mulube
- PATH-Malaria Control and Elimination Partnership in Africa (MACEPA), National Malaria Elimination Centre, Lusaka, Zambia
| | | | - Jessica L Schue
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - William J Moss
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Qixin He
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.
| | - Giovanna Carpi
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA; Purdue Institute of Inflammation, Immunology and Infectious Disease, West Lafayette, IN, USA.
| |
Collapse
|
10
|
Amen A, Yoo R, Fabra-García A, Bolscher J, Stone WJR, Bally I, Dergan-Dylon S, Kucharska I, de Jong RM, de Bruijni M, Bousema T, Richter King C, MacGill RS, Sauerwein RW, Julien JP, Poignard P, Jore MM. Target-agnostic identification of human antibodies to Plasmodium falciparum sexual forms reveals cross stage recognition of glutamate-rich repeats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.03.565335. [PMID: 37961136 PMCID: PMC10635103 DOI: 10.1101/2023.11.03.565335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Circulating sexual stages of Plasmodium falciparum (Pf) can be transmitted from humans to mosquitoes, thereby furthering the spread of malaria in the population. It is well established that antibodies (Abs) can efficiently block parasite transmission. In search for naturally acquired Ab targets on sexual stages, we established an efficient method for target-agnostic single B cell activation followed by high-throughput selection of human monoclonal antibodies (mAbs) reactive to sexual stages of Pf in the form of gamete and gametocyte extract. We isolated mAbs reactive against a range of Pf proteins including well-established targets Pfs48/45 and Pfs230. One mAb, B1E11K, was cross-reactive to various proteins containing glutamate-rich repetitive elements expressed at different stages of the parasite life cycle. A crystal structure of two B1E11K Fab domains in complex with its main antigen, RESA, expressed on asexual blood stages, showed binding of B1E11K to a repeating epitope motif in a head-to-head conformation engaging in affinity-matured homotypic interactions. Thus, this mode of recognition of Pf proteins, previously described only for PfCSP, extends to other repeats expressed across various stages. The findings augment our understanding of immune-pathogen interactions to repeating elements of the Plasmodium parasite proteome and underscore the potential of the novel mAb identification method used to provide new insights into the natural humoral immune response against Pf . Impact Statement A naturally acquired human monoclonal antibody recognizes proteins expressed at different stages of the Plasmodium falciparum lifecycle through affinity-matured homotypic interactions with glutamate-rich repeats.
Collapse
|
11
|
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
|
12
|
Salinas ND, Ma R, McAleese H, Ouahes T, Long CA, Miura K, Lambert LE, Tolia NH. A Self-Assembling Pfs230D1-Ferritin Nanoparticle Vaccine Has Potent and Durable Malaria Transmission-Reducing Activity. Vaccines (Basel) 2024; 12:546. [PMID: 38793797 PMCID: PMC11125772 DOI: 10.3390/vaccines12050546] [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: 04/12/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Malaria is caused by eukaryotic protozoan parasites of the genus Plasmodium. There are 249 million new cases and 608,000 deaths annually, and new interventions are desperately needed. Malaria vaccines can be divided into three categories: liver stage, blood stage, or transmission-blocking vaccines. Transmission-blocking vaccines prevent the transmission of disease by the mosquito vector from one human to another. Pfs230 is one of the leading transmission-blocking vaccine antigens for malaria. Here, we describe the development of a 24-copy self-assembling nanoparticle vaccine comprising domain 1 of Pfs230 genetically fused to H. pylori ferritin. The single-component Pfs230D1-ferritin construct forms a stable and homogenous 24-copy nanoparticle with good production yields. The nanoparticle is highly immunogenic, as two low-dose vaccinations of New Zealand White rabbits elicited a potent and durable antibody response with high transmission-reducing activity when formulated in two distinct adjuvants suitable for translation to human use. This single-component 24-copy Pfs230D1-ferritin nanoparticle vaccine has the potential to improve production pipelines and the cost of manufacturing a potent and durable transmission-blocking vaccine for malaria control.
Collapse
Affiliation(s)
- Nichole D. Salinas
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (N.D.S.)
| | - Rui Ma
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (N.D.S.)
| | - Holly McAleese
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tarik Ouahes
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Carole A. Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Lynn E. Lambert
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Niraj H. Tolia
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (N.D.S.)
| |
Collapse
|
13
|
Ciubotariu II, Broyles BK, Xie S, Thimmapuram J, Mwenda MC, Mambwe B, Mulube C, Matoba J, Schue JL, Moss WJ, Bridges DJ, He Q, Carpi G. Diversity and selection analyses identify transmission-blocking antigens as the optimal vaccine candidates in Plasmodium falciparum. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.11.24307175. [PMID: 38766239 PMCID: PMC11100930 DOI: 10.1101/2024.05.11.24307175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Background A highly effective vaccine for malaria remains an elusive target, at least in part due to the under-appreciated natural parasite variation. This study aimed to investigate genetic and structural variation, and immune selection of leading malaria vaccine candidates across the Plasmodium falciparum's life cycle. Methods We analyzed 325 P. falciparum whole genome sequences from Zambia, in addition to 791 genomes from five other African countries available in the MalariaGEN Pf3k Rdatabase. Ten vaccine antigens spanning three life-history stages were examined for genetic and structural variations, using population genetics measures, haplotype network analysis, and 3D structure selection analysis. Findings Among the ten antigens analyzed, only three in the transmission-blocking vaccine category display P. falciparum 3D7 as the dominant haplotype. The antigens AMA1, CSP, MSP119 and CelTOS, are much more diverse than the other antigens, and their epitope regions are under moderate to strong balancing selection. In contrast, Rh5, a blood stage antigen, displays low diversity yet slightly stronger immune selection in the merozoite-blocking epitope region. Except for CelTOS, the transmission-blocking antigens Pfs25, Pfs48/45, Pfs230, Pfs47, and Pfs28 exhibit minimal diversity and no immune selection in epitopes that induce strain-transcending antibodies, suggesting potential effectiveness of 3D7-based vaccines in blocking transmission. Interpretations These findings offer valuable insights into the selection of optimal vaccine candidates against P. falciparum. Based on our results, we recommend prioritizing conserved merozoite antigens and transmission-blocking antigens. Combining these antigens in multi-stage approaches may be particularly promising for malaria vaccine development initiatives. Funding Purdue Department of Biological Sciences; Puskas Memorial Fellowship; National Institute of Allergy and Infectious Diseases (U19AI089680).
Collapse
Affiliation(s)
- Ilinca I. Ciubotariu
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Bradley K. Broyles
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Shaojun Xie
- Bioinformatics Core, Purdue University, West Lafayette, Indiana, USA
| | | | - Mulenga C. Mwenda
- PATH-Malaria Control and Elimination Partnership in Africa (MACEPA), National Malaria Elimination Centre, Lusaka, Zambia
| | - Brenda Mambwe
- PATH-Malaria Control and Elimination Partnership in Africa (MACEPA), National Malaria Elimination Centre, Lusaka, Zambia
| | - Conceptor Mulube
- PATH-Malaria Control and Elimination Partnership in Africa (MACEPA), National Malaria Elimination Centre, Lusaka, Zambia
| | | | - Jessica L. Schue
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - William J. Moss
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | | | - Qixin He
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Giovanna Carpi
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute of Inflammation, Immunology and Infectious Disease, West Lafayette, Indiana, USA
| |
Collapse
|
14
|
Alkema M, Smit MJ, Marin-Mogollon C, Totté K, Teelen K, van Gemert GJ, van de Vegte-Bolmer M, Mordmüller BG, Reimer JM, Lövgren-Bengtsson KL, Sauerwein RW, Bousema T, Plieskatt J, Theisen M, Jore MM, McCall MBB. A Pfs48/45-based vaccine to block Plasmodium falciparum transmission: phase 1, open-label, clinical trial. BMC Med 2024; 22:170. [PMID: 38649867 PMCID: PMC11036667 DOI: 10.1186/s12916-024-03379-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND The stalling global progress in malaria control highlights the need for novel tools for malaria elimination, including transmission-blocking vaccines. Transmission-blocking vaccines aim to induce human antibodies that block parasite development in the mosquito and mosquitoes becoming infectious. The Pfs48/45 protein is a leading Plasmodium falciparum transmission-blocking vaccine candidate. The R0.6C fusion protein, consisting of Pfs48/45 domain 3 (6C) and the N-terminal region of P. falciparum glutamate-rich protein (R0), has previously been produced in Lactococcus lactis and elicited functional antibodies in rodents. Here, we assess the safety and transmission-reducing efficacy of R0.6C adsorbed to aluminium hydroxide with and without Matrix-M™ adjuvant in humans. METHODS In this first-in-human, open-label clinical trial, malaria-naïve adults, aged 18-55 years, were recruited at the Radboudumc in Nijmegen, the Netherlands. Participants received four intramuscular vaccinations on days 0, 28, 56 and 168 with either 30 µg or 100 µg of R0.6C and were randomised for the allocation of one of the two different adjuvant combinations: aluminium hydroxide alone, or aluminium hydroxide combined with Matrix-M1™ adjuvant. Adverse events were recorded from inclusion until 84 days after the fourth vaccination. Anti-R0.6C and anti-6C IgG titres were measured by enzyme-linked immunosorbent assay. Transmission-reducing activity of participants' serum and purified vaccine-specific immunoglobulin G was assessed by standard membrane feeding assays using laboratory-reared Anopheles stephensi mosquitoes and cultured P. falciparum gametocytes. RESULTS Thirty-one participants completed four vaccinations and were included in the analysis. Administration of all doses was safe and well-tolerated, with one related grade 3 adverse event (transient fever) and no serious adverse events occurring. Anti-R0.6C and anti-6C IgG titres were similar between the 30 and 100 µg R0.6C arms, but higher in Matrix-M1™ arms. Neat participant sera did not induce significant transmission-reducing activity in mosquito feeding experiments, but concentrated vaccine-specific IgGs purified from sera collected two weeks after the fourth vaccination achieved up to 99% transmission-reducing activity. CONCLUSIONS R0.6C/aluminium hydroxide with or without Matrix-M1™ is safe, immunogenic and induces functional Pfs48/45-specific transmission-blocking antibodies, albeit at insufficient serum concentrations to result in transmission reduction by neat serum. Future work should focus on identifying alternative vaccine formulations or regimens that enhance functional antibody responses. TRIAL REGISTRATION The trial is registered with ClinicalTrials.gov under identifier NCT04862416.
Collapse
Affiliation(s)
- M Alkema
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - M J Smit
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - C Marin-Mogollon
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - K Totté
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - K Teelen
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - G J van Gemert
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - M van de Vegte-Bolmer
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - B G Mordmüller
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | | | - R W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
- Present Address: TropIQ Health Sciences, Nijmegen, the Netherlands
| | - T Bousema
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - J Plieskatt
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - M Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - M M Jore
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - M B B McCall
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
15
|
Plieskatt J, Bang P, Wood GK, Naghizadeh M, Singh SK, Jore MM, Theisen M. Clinical formulation development of Plasmodium falciparum malaria vaccine candidates based on Pfs48/45, Pfs230, and PfCSP. Vaccine 2024; 42:1980-1992. [PMID: 38388238 DOI: 10.1016/j.vaccine.2024.02.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/09/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024]
Abstract
Two malaria transmission-blocking vaccine (TBV) candidates, R0.6C and ProC6C, have completed preclinical development including the selection of adjuvants, Alhydrogel® with or without the saponin based adjuvant Matrix-M™. Here, we report on the final drug product (formulation) design of R0.6C and ProC6C and evaluate their safety and biochemical stability in preparation for preclinical and clinical pharmacy handling. The point-of-injection stability studies demonstrated that both the R0.6C and ProC6C antigens are stable on Alhydrogel in the presence or absence of Matrix-M for up to 24 h at room temperature. As this is the first study to combine Alhydrogel and Matrix-M for clinical use, we also evaluated their potential interactions. Matrix-M adsorbs to Alhydrogel, while not displacing the > 95 % adsorbed protein. The R0.6C and ProC6C formulations were found to be safe and well tolerated in repeated dose toxicity studies in rabbits generating high levels of functional antibodies that blocked infection of mosquitoes. Further, the R0.6C and ProC6C drug products were found to be stable for minimally 24 months when stored at 2-8 °C, with studies ongoing through 36 months. Together, this data demonstrates the safety and suitability of the L. lactis expression system as well as supports the clinical testing of the R0.6C and ProC6C malaria vaccine candidates in First-In-Human clinical trials.
Collapse
Affiliation(s)
- Jordan Plieskatt
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Bang
- Department of Vaccine Development, Statens Serum Institut, Copenhagen, Denmark
| | - Grith Krøyer Wood
- Department of Vaccine Development, Statens Serum Institut, Copenhagen, Denmark
| | - Mohammad Naghizadeh
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Susheel K Singh
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Matthijs M Jore
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
16
|
Beeson JG, Chan JA. A step forward for Plasmodium falciparum malaria transmission-blocking vaccines. THE LANCET. INFECTIOUS DISEASES 2023; 23:1210-1212. [PMID: 37499677 DOI: 10.1016/s1473-3099(23)00288-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 05/02/2023] [Indexed: 07/29/2023]
Affiliation(s)
- James G Beeson
- Burnet Institute, Melbourne, VIC, Australia; Department of Microbiology, Monash University, Melbourne, VIC, Australia; Departments of Infectious Diseases and Medicine, The University of Melbourne, Melbourne, VIC, Australia.
| | - Jo-Anne Chan
- Burnet Institute, Melbourne, VIC, Australia; Department of Immunology, Monash University, Melbourne, VIC, Australia; Departments of Infectious Diseases and Medicine, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
17
|
Dickey TH, Tolia NH. Designing an effective malaria vaccine targeting Plasmodium vivax Duffy-binding protein. Trends Parasitol 2023; 39:850-858. [PMID: 37481347 PMCID: PMC11099547 DOI: 10.1016/j.pt.2023.06.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/24/2023]
Abstract
Malaria caused by the Plasmodium vivax parasite is a major global health burden. Immunity against blood-stage infection reduces parasitemia and disease severity. Duffy-binding protein (DBP) is the primary parasite protein responsible for the invasion of red blood cells and it is a leading subunit vaccine candidate. An effective vaccine, however, is still lacking despite decades of interest in DBP as a vaccine candidate. This review discusses the reasons for targeting DBP, the challenges associated with developing a vaccine, and modern structural vaccinology methods that could be used to create an effective DBP vaccine. Next-generation DBP vaccines have the potential to elicit a broadly protective immune response and provide durable and potent protection from P. vivax malaria.
Collapse
Affiliation(s)
- Thayne H Dickey
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20894, USA
| | - Niraj H Tolia
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20894, USA.
| |
Collapse
|
18
|
Shukla N, Tang WK, Coelho CH, Long CA, Healy SA, Sagara I, Miura K, Duffy PE, Tolia NH. A human antibody epitope map of the malaria vaccine antigen Pfs25. NPJ Vaccines 2023; 8:108. [PMID: 37542029 PMCID: PMC10403551 DOI: 10.1038/s41541-023-00712-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 07/12/2023] [Indexed: 08/06/2023] Open
Abstract
Pfs25 is a leading antigen for a malaria transmission-blocking vaccine and shows moderate transmission-blocking activity and induction of rapidly decreasing antibody titers in clinical trials. A comprehensive definition of all transmission-reducing epitopes of Pfs25 will inform structure-guided design to enhance Pfs25-based vaccines, leading to potent transmission-blocking activity. Here, we compiled a detailed human antibody epitope map comprising epitope binning data and structures of multiple human monoclonal antibodies, including three new crystal structures of Pfs25 in complex with transmission-reducing antibodies from Malian volunteers immunized with Pfs25 conjugated to EPA and adjuvanted with AS01. These structures revealed additional epitopes in Pfs25 capable of reducing transmission and expanded this characterization to malaria-exposed humans. This work informs immunogen design to focus the antibody response to transmission-reducing epitopes of Pfs25, enabling development of more potent transmission-blocking vaccines for malaria.
Collapse
Affiliation(s)
- Niharika Shukla
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Wai Kwan Tang
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Camila H Coelho
- Pathogenesis and Immunity Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Sara A Healy
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Issaka Sagara
- Malaria Research and Training Center, University of Sciences, Techniques, and Technology, Bamako, Mali
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Patrick E Duffy
- Pathogenesis and Immunity Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Niraj H Tolia
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA.
| |
Collapse
|
19
|
X marks the shot against malaria. Immunity 2023; 56:234-236. [PMID: 36792569 DOI: 10.1016/j.immuni.2023.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
The development of a transmission-blocking vaccine (TBV) against malaria is hampered by poor understanding of functional antibody responses. In this issue of Immunity, Fabra-Garcia et al., Ivanochko et al., and Tang et al. isolate human monoclonal antibodies against the two most promising TBV candidates, Pfs48/45 and Pfs230, and map the epitopes responsible for potent transmission-reducing activity.
Collapse
|