1
|
Hickey JM, Sharma N, Fairlamb M, Doering J, Adewunmi Y, Prieto K, Costa G, Wizel B, Levashina EA, Mantis NJ, Julien JP, Joshi SB, Volkin DB. PfCSP-ferritin nanoparticle malaria vaccine antigen formulated with aluminum-salt and CpG 1018® adjuvants: Preformulation characterization, antigen-adjuvant interactions, and mouse immunogenicity studies. Hum Vaccin Immunother 2025; 21:2460749. [PMID: 39903060 PMCID: PMC11796538 DOI: 10.1080/21645515.2025.2460749] [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: 11/15/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025] Open
Abstract
Circumsporozite protein (CSP), the most abundant surface protein in parasitic Plasmodium falciparum (Pf) sporozoite and an attractive target for malaria vaccine design, has been shown to induce protective humoral response in humans. In this work, we characterized and formulated a promising recombinant PfCSP immunogen (155) candidate consisting of two PfCSP epitopes (i.e. junction, NANP repeat) fused to H. pylori apoferritin forming a 24-mer nanoparticle. In addition, two N-linked glycans were engineered to mitigate possible anti-apoferritin immune responses, and a universal T-cell epitope was included to further enhance immunogenicity. Physicochemical characterization of the 155 antigen was performed including primary structure, post-translational modifications, conformational stability, and particle size. A competitive ELISA was developed to assess antigen binding to a PfCSP-specific mAb. The in vitro antigenicity of the 155 antigen was measured upon formulation with adjuvants, including aluminum-salts (i.e. AlhydrogelTM, Adju-PhosTM) and the TLR-9 agonist CpG 1018®, when freshly combined and after storage at different temperatures over 3 months. The in vivo immunological impact of various adjuvanted formulations of the 155 antigen was investigated in mice. The results support the formulation of 155 with AlhydrogelTM + CpG 1018® adjuvants as a promising recombinant malaria vaccine candidate from both a pharmaceutical and immunological perspective.
Collapse
MESH Headings
- Animals
- Malaria Vaccines/immunology
- Malaria Vaccines/administration & dosage
- Malaria Vaccines/chemistry
- Adjuvants, Immunologic/administration & dosage
- Nanoparticles/administration & dosage
- Nanoparticles/chemistry
- Protozoan Proteins/immunology
- Protozoan Proteins/genetics
- Oligodeoxyribonucleotides/administration & dosage
- Adjuvants, Vaccine/administration & dosage
- Immunogenicity, Vaccine
- Plasmodium falciparum/immunology
- Mice
- Female
- Antibodies, Protozoan/blood
- Antigens, Protozoan/immunology
- Antigens, Protozoan/genetics
- Mice, Inbred BALB C
- Ferritins/immunology
- Ferritins/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Malaria, Falciparum/prevention & control
- Aluminum Hydroxide/administration & dosage
Collapse
Affiliation(s)
- John M. Hickey
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Nitya Sharma
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Max Fairlamb
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Jennifer Doering
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Yetunde Adewunmi
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Katherine Prieto
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, ON, Canada
| | - Giulia Costa
- Vector Biology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Benjamin Wizel
- Head of External Research and Development, Dynavax Technologies Corporation, Emeryville, CA, USA
| | - Elena A. Levashina
- Vector Biology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Nicholas J. Mantis
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, ON, Canada
- Departments of Biochemistry and Immunology, University of Toronto, Toronto, ON, Canada
| | - Sangeeta B. Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - David B. Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
2
|
Ukegbu CV, Mohamed M, Hoermann A, Qin Y, Kweyamba PA, Lwetoijera DW, Windbichler N, Moore S, Christophides GK, Vlachou D. Nanobody-mediated targeting of Plasmodium falciparum PfPIMMS43 can block malaria transmission in mosquitoes. Commun Biol 2025; 8:683. [PMID: 40301628 PMCID: PMC12041390 DOI: 10.1038/s42003-025-08033-8] [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: 12/04/2024] [Accepted: 04/02/2025] [Indexed: 05/01/2025] Open
Abstract
The transition from ookinete to oocyst is a critical step in the Plasmodium falciparum lifecycle and an important target for malaria transmission-blocking strategies. PfPIMMS43, a surface protein of P. falciparum ookinetes and sporozoites, is critical for this transition and aids the parasite in evading mosquito immune responses. Previous studies demonstrated that polyclonal PfPIMMS43 antibodies reduced P. falciparum infection in Anopheles mosquitoes. Here, building on these findings, we have developed high-affinity single-domain VHH antibodies (nanobodies) derived from llama heavy-chain-only antibodies. We have shown that these nanobodies bind both recombinant and endogenous PfPIMMS43 produced by P. falciparum ookinetes in the mosquito midgut. Importantly, they significantly reduce infection intensity and prevalence of laboratory and field strains of P. falciparum in An. coluzzii and An. gambiae, respectively. Epitope mapping has revealed that the nanobodies target conserved regions in the second half of PfPIMMS43, with homology modelling confirming epitope accessibility. These findings establish PfPIMMS43 as a promising transmission-blocking target. To enhance malaria control and elimination efforts, we propose an innovative strategy in which genetically modified mosquitoes express PfPIMMS43-specific nanobodies in their midguts and spread this trait in wild mosquito populations via gene drive technology.
Collapse
Affiliation(s)
| | - Mgeni Mohamed
- Environmental Health and Ecological Sciences, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Astrid Hoermann
- Department of Life Sciences, Imperial College London, London, UK
| | - Yuyan Qin
- Department of Life Sciences, Imperial College London, London, UK
| | - Prisca A Kweyamba
- Environmental Health and Ecological Sciences, Ifakara Health Institute, Bagamoyo, Tanzania
| | | | | | - Sarah Moore
- Environmental Health and Ecological Sciences, Ifakara Health Institute, Bagamoyo, Tanzania
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | | | - Dina Vlachou
- Department of Life Sciences, Imperial College London, London, UK.
| |
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
|
Duffy PE, Gorres JP, Healy SA, Fried M. Malaria vaccines: a new era of prevention and control. Nat Rev Microbiol 2024; 22:756-772. [PMID: 39025972 DOI: 10.1038/s41579-024-01065-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2024] [Indexed: 07/20/2024]
Abstract
Malaria killed over 600,000 people in 2022, a death toll that has not improved since 2015. Additionally, parasites and mosquitoes resistant to existing interventions are spreading across Africa and other regions. Vaccines offer hope to reduce the mortality burden: the first licensed malaria vaccines, RTS,S and R21, will be widely deployed in 2024 and should substantially reduce childhood deaths. In this Review, we provide an overview of the malaria problem and the Plasmodium parasite, then describe the RTS,S and R21 vaccines (the first vaccines for any human parasitic disease), summarizing their benefits and limitations. We explore next-generation vaccines designed using new knowledge of malaria pathogenesis and protective immunity, which incorporate antigens and platforms to elicit effective immune responses against different parasite stages in human or mosquito hosts. We describe a decision-making process that prioritizes malaria vaccine candidates for development in a resource-constrained environment. Future vaccines might improve upon the protective efficacy of RTS,S or R21 for children, or address the wider malaria scourge by preventing pregnancy malaria, reducing the burden of Plasmodium vivax or accelerating malaria elimination.
Collapse
Affiliation(s)
- Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - J Patrick Gorres
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sara A Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michal Fried
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
5
|
Nourani L, Ayoub Meigouni M, Afzali S, Zargar M, Pourhashem Z, Yousefi H, Sani JJ, Vand-Rajabpour H, Pirahmadi S, Raz A, Abouie Mehrizi A. Cd loop fusion enhances the immunogenicity and the potential transmission blocking activity of Plasmodium falciparum generative cell specific 1 (GCS1) antigen. Biochem Biophys Res Commun 2024; 733:150599. [PMID: 39208643 DOI: 10.1016/j.bbrc.2024.150599] [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: 06/15/2024] [Revised: 08/10/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
TBVs are suggested to inhibit parasite transmission from humans to Anopheles mosquitoes. For the transmission of Plasmodium parasite, a variety of factors are included in gametes fusion phase. In this step, conserved male-specific generative cell specific 1 antigen is necessary for fusion of cytoplasmic membranes of micro- and macro-gametocytes and zygot formation. The partial blocking activities of elicited antibodies against either the HAP2-GCS1 domain or the cd loop of this antigen have been recorded to hinder the transmission of Plasmodium species in Anopheles mid-gut. Thus, the objective of the present study was to investigate if the cd loop-fusion can enhance the quantity and quality of humoral and cellular immune responses against Plasmodium falciparum GCS1 in comparison to non-fusion antigen (without cd loop), in the adjuvanted and non-adjuvanted mouse groups. The immunogenicity of two constructs of P. falciparum generative cell specific 1 antigen, a fusion protein composed of cd loop and HAP2-GCS1 domain (cd-HAP) and another recombinant PfGCS1 containing solo HAP2-GCS1 domain (HAP2) were assessed to impede Plasmodium gametocytes integration before zygote formation. The antibodies profiling, titer, and avidity of induced antibodies were measured by the immunized mice sera, and the released cytokines (IL-5, TNF, and INF-γ) were analyzed in the supernatants of stimulated splenocytes. Furthermore, the inhibitory potency of the elicited antibodies against HAP2 and cd-HAP was measured during oocyst development by Standard Membrane Feeding Assay (SMFA). The comparative results in the present study showed the higher titer of IgG antibodies and IgG2a subclass, avidity, and transmission-reducing activity (TRA = 72.5 %) when mice were immunized by cd-HAP rather than HAP2. Moreover, our findings confirmed intensified Th1-directed immune responses in group 4 received cd-HAP/Poly(I:C). These findings declared the potential ability of cd loop fusion (cd-HAP) to upsurge humoral and cellular immune responses. However, the immune responses may switch to stronger Th1-type using alternative formulations. Explicitly, the cd-HAP-based vaccine may enhance the overall efficiency of immune responses and present a promising implementation in aiming malaria transmission.
Collapse
Affiliation(s)
- Leila Nourani
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Masoumeh Ayoub Meigouni
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Shima Afzali
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Mostafa Zargar
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Zeinab Pourhashem
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Hemn Yousefi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Jafar J Sani
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Hediye Vand-Rajabpour
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Abbasali Raz
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Akram Abouie Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
6
|
Cao Y, Hayashi CTH, Araujo MDS, Tripathi AK, Andrade AO, Medeiros JF, Vinetz J, Kumar N. Evaluation of combination vaccines targeting transmission of Plasmodium falciparum and P. vivax. Vaccine 2024; 42:126140. [PMID: 39033079 PMCID: PMC11338703 DOI: 10.1016/j.vaccine.2024.07.041] [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: 07/02/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
Transmission-blocking vaccines interrupting malaria transmission within mosquitoes represent an ideal public health tool to eliminate malaria at the population level. Plasmodium falciparum and P. vivax account for more than 90% of the global malaria burden, co-endemic in many regions of the world. P25 and P48/45 are two leading candidates for both species and have shown promising transmission-blocking activity in preclinical and clinical studies. However, neither of these target antigens as individual vaccines has induced complete transmission inhibition in mosquitoes. In this study, we assessed immunogenicity of combination vaccines based on P25 and P48/45 using a DNA vaccine platform to broaden vaccine specificity against P. falciparum and P. vivax. Individual DNA vaccines encoding Pvs25, Pfs25, Pvs48/45 and Pfs48/45, as well as various combinations including (Pvs25 + Pvs48/45), (Pfs25 + Pfs48/45), (Pvs25 + Pfs25), and (Pvs48/45 + Pfs48/45), were evaluated in mice using in vivo electroporation. Potent antibody responses were induced in mice immunized with individual and combination DNA vaccines, and specific antibody responses were not compromised when combinations of DNA vaccines were evaluated against individual DNA vaccines. The anti-Pvs25 IgG from individual and combination groups revealed concentration-dependent transmission-reducing activity (TRA) in direct membrane feeding assays (DMFA) using blood from P. vivax-infected donors in Brazil and independently in ex vivo MFA using Pvs25-transgenic P. berghei. Similarly, anti-Pfs25 and anti-Pfs48/45 IgGs from mice immunized with Pfs25 and Pfs48/45 DNA vaccines individually and in various combinations revealed antibody dose-dependent TRA in standard membrane feeding assays (SMFA) using culture-derived P. falciparum gametocytes. However, antibodies induced by immunization with Pvs48/45 DNA vaccines were ineffective in DMFA and require further vaccine construct optimization, considering the possibility of induction of both transmission-blocking and transmission-enhancing antibodies revealed by competition ELISA. These studies provide a rationale for combining multiple antigens to simultaneously target transmission of malaria caused by P. falciparum and P. vivax.
Collapse
MESH Headings
- Malaria Vaccines/immunology
- Malaria Vaccines/administration & dosage
- Animals
- Malaria, Falciparum/prevention & control
- Malaria, Falciparum/transmission
- Malaria, Falciparum/immunology
- Plasmodium falciparum/immunology
- Plasmodium falciparum/genetics
- Plasmodium vivax/immunology
- Plasmodium vivax/genetics
- Malaria, Vivax/prevention & control
- Malaria, Vivax/transmission
- Malaria, Vivax/immunology
- Mice
- Vaccines, DNA/immunology
- Vaccines, DNA/administration & dosage
- Antibodies, Protozoan/immunology
- Antibodies, Protozoan/blood
- Female
- Vaccines, Combined/immunology
- Vaccines, Combined/administration & dosage
- Antigens, Protozoan/immunology
- Antigens, Protozoan/genetics
- Protozoan Proteins/immunology
- Protozoan Proteins/genetics
- Mice, Inbred BALB C
- Humans
Collapse
Affiliation(s)
- Yi Cao
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington D.C., USA
| | - Clifford T H Hayashi
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington D.C., USA
| | - Maisa da Silva Araujo
- Plataforma de Produção e Infecção de Vetores da Malária, Laboratório de Entomologia - FIOCRUZ RO, Rua da Beira 7671, CEP 76812-245 Porto Velho RO, Brazil; Programa de Pós-Graduação em Saúde Pública, Faculdade de Saúde Pública, Universidade Federal de São Paulo, São Paulo 01246-904, SP, Brazil
| | - Abhai K Tripathi
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Alice Oliveira Andrade
- Plataforma de Produção e Infecção de Vetores da Malária, Laboratório de Entomologia - FIOCRUZ RO, Rua da Beira 7671, CEP 76812-245 Porto Velho RO, Brazil; Programa de Pós-Graduação em Saúde Pública, Faculdade de Saúde Pública, Universidade Federal de São Paulo, São Paulo 01246-904, SP, Brazil
| | - Jansen Fernandes Medeiros
- Plataforma de Produção e Infecção de Vetores da Malária, Laboratório de Entomologia - FIOCRUZ RO, Rua da Beira 7671, CEP 76812-245 Porto Velho RO, Brazil; Programa de Pós-Graduação em Biologia Experimental, Fundação Universidade Federal de Rondônia, Fiocruz Rondônia 76812-245, Brazil
| | - Joseph Vinetz
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA; Department of Cellular and Molecular Sciences, Faculty of Sciences, and Alexander von Humboldt Institute of Tropical Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Nirbhay Kumar
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington D.C., USA.
| |
Collapse
|
7
|
Coelho CH, Marquez S, Nguemwo Tentokam BC, Berhe AD, Miura K, Rao VN, Long CA, Doumbo OK, Sagara I, Healy S, Kleinstein SH, Duffy PE. Antibody gene features associated with binding and functional activity in malaria vaccine-derived human mAbs. NPJ Vaccines 2024; 9:144. [PMID: 39127706 PMCID: PMC11316794 DOI: 10.1038/s41541-024-00929-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
The impact of adjuvants on malaria vaccine-induced antibody repertoire is poorly understood. Here, we characterize the impact of two adjuvants, Alhydrogel® and AS01, on antibody clonotype diversity, binding and function, post malaria vaccination. We expressed 132 recombinant anti-Pfs230D1 human monoclonal antibodies (mAbs) from participants immunized with malaria transmission-blocking vaccine Pfs230D1, formulated with either Alhydrogel® or AS01. Anti-Pfs230D1 mAbs generated by Alhydrogel® formulation showed higher binding frequency to Pfs230D1 compared to AS01 formulation, although the frequency of functional mAbs was similar between adjuvant groups. Overall, the AS01 formulation induced anti-Pfs230D1 functional antibodies from a broader array of germline sequences versus the Alhydrogel® formulation. All mAbs using IGHV1-69 gene from the Alhydrogel® cohort bound to recombinant Pfs230D1, but did not block parasite transmission to mosquitoes, similar to the IGHV1-69 mAbs isolated from the AS01 cohort. These findings may help inform vaccine design and adjuvant selection for immunization with Plasmodium antigens.
Collapse
Affiliation(s)
- Camila H Coelho
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- C-VARPP- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Immunology Precision Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Susanna Marquez
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Bergeline C Nguemwo Tentokam
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Anne D Berhe
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, USA
| | - Vishal N Rao
- C-VARPP- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, USA
| | - Ogobara K Doumbo
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Issaka Sagara
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Sara Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Steven H Kleinstein
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, 06511, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
8
|
Scaria PV, Roth N, Schwendt K, Muratova OV, Alani N, Lambert LE, Barnafo EK, Rowe CG, Zaidi IU, Rausch KM, Narum DL, Petsch B, Duffy PE. mRNA vaccines expressing malaria transmission-blocking antigens Pfs25 and Pfs230D1 induce a functional immune response. NPJ Vaccines 2024; 9:9. [PMID: 38184666 PMCID: PMC10771442 DOI: 10.1038/s41541-023-00783-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/22/2023] [Indexed: 01/08/2024] Open
Abstract
Malaria transmission-blocking vaccines (TBV) are designed to inhibit the sexual stage development of the parasite in the mosquito host and can play a significant role in achieving the goal of malaria elimination. Preclinical and clinical studies using protein-protein conjugates of leading TBV antigens Pfs25 and Pfs230 domain 1 (Pfs230D1) have demonstrated the feasibility of TBV. Nevertheless, other promising vaccine platforms for TBV remain underexplored. The recent success of mRNA vaccines revealed the potential of this technology for infectious diseases. We explored the mRNA platform for TBV development. mRNA constructs of Pfs25 and Pfs230D1 variously incorporating signal peptides (SP), GPI anchor, and Trans Membrane (TM) domain were assessed in vitro for antigen expression, and selected constructs were evaluated in mice. Only mRNA constructs with GPI anchor or TM domain that resulted in high cell surface expression of the antigens yielded strong immune responses in mice. These mRNA constructs generated higher transmission-reducing functional activity versus the corresponding alum-adjuvanted protein-protein conjugates used as comparators. Pfs25 mRNA with GPI anchor or TM maintained >99% transmission reducing activity through 126 days, the duration of the study, demonstrating the potential of mRNA platform for TBV.
Collapse
Affiliation(s)
- Puthupparampil V Scaria
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | | | | | - Olga V Muratova
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | - Nada Alani
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | - Lynn E Lambert
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | - Emma K Barnafo
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | - Christopher G Rowe
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | - Irfan U Zaidi
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | - Kelly M Rausch
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | - David L Narum
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | | | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA.
| |
Collapse
|
9
|
Bansal GP, Kumar N. Immune mechanisms targeting malaria transmission: opportunities for vaccine development. Expert Rev Vaccines 2024; 23:645-654. [PMID: 38888098 PMCID: PMC11472754 DOI: 10.1080/14760584.2024.2369583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
INTRODUCTION Malaria continues to remain a major global health problem with nearly a quarter of a billion clinical cases and more than 600,000 deaths in 2022. There has been significant progress toward vaccine development, however, poor efficacy of approved vaccines requiring multiple immunizing doses emphasizes the need for continued efforts toward improved vaccines. Progress to date, nonetheless, has provided impetus for malaria elimination. AREAS COVERED In this review we will focus on diverse immune mechanisms targeting gametocytes in the human host and gametocyte-mediated malaria transmission via the mosquito vector. EXPERT OPINION To march toward the goal of malaria elimination it will be critical to target the process of malaria transmission by mosquitoes, mediated exclusively by the sexual stages, i.e. male, and female gametocytes, ingested from infected vertebrate host. Studies over several decades have established antigens in the parasite sexual stages developing in the mosquito midgut as attractive targets for the development of transmission blocking vaccines (TBVs). Immune clearance of gametocytes in the vertebrate host can synergize with TBVs and directly aid in maintaining effective transmission reducing immune potential.
Collapse
Affiliation(s)
- Geetha P. Bansal
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70122, USA
| | - Nirbhay Kumar
- Department of Global Health, The Milken Institute School of Public Health, George Washington University, Washington DC, 20052, USA
| |
Collapse
|
10
|
Doritchamou J, Nielsen MA, Chêne A, Viebig NK, Lambert LE, Sander AF, Semblat JP, Hundt S, Orr-Gonzalez S, Janitzek CM, Spiegel AJ, Clemmensen SB, Thomas ML, Nason MC, Snow-Smith M, Barnafo EK, Shiloach J, Chen BB, Nadakal S, Highsmith K, Ouahes T, Conteh S, Sharma A, Torano H, Butler B, Reiter K, Rausch KM, Scaria PV, Anderson C, Narum DL, Salanti A, Fried M, Theander TG, Gamain B, Duffy PE. Aotus nancymaae model predicts human immune response to the placental malaria vaccine candidate VAR2CSA. Lab Anim (NY) 2023; 52:315-323. [PMID: 37932470 PMCID: PMC10689237 DOI: 10.1038/s41684-023-01274-2] [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: 09/13/2022] [Accepted: 09/27/2023] [Indexed: 11/08/2023]
Abstract
Placental malaria vaccines (PMVs) are being developed to prevent severe sequelae of placental malaria (PM) in pregnant women and their offspring. The leading candidate vaccine antigen VAR2CSA mediates parasite binding to placental receptor chondroitin sulfate A (CSA). Despite promising results in small animal studies, recent human trials of the first two PMV candidates (PAMVAC and PRIMVAC) generated limited cross-reactivity and cross-inhibitory activity to heterologous parasites. Here we immunized Aotus nancymaae monkeys with three PMV candidates (PAMVAC, PRIMVAC and ID1-ID2a_M1010) adjuvanted with Alhydrogel, and exploited the model to investigate boosting of functional vaccine responses during PM episodes as well as with nanoparticle antigens. PMV candidates induced high levels of antigen-specific IgG with significant cross-reactivity across PMV antigens by enzyme-linked immunosorbent assay. Conversely, PMV antibodies recognized native VAR2CSA and blocked CSA adhesion of only homologous parasites and not of heterologous parasites. PM episodes did not significantly boost VAR2CSA antibody levels or serum functional activity; nanoparticle and monomer antigens alike boosted serum reactivity but not functional activities. Overall, PMV candidates induced functional antibodies with limited heterologous activity in Aotus monkeys, similar to responses reported in humans. The Aotus model appears suitable for preclinical downselection of PMV candidates and assessment of antibody boosting by PM episodes.
Collapse
Affiliation(s)
- Justin Doritchamou
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Morten A Nielsen
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Arnaud Chêne
- Université Paris Cité and Université des Antilles, INSERM, BIGR, Paris, France
| | - Nicola K Viebig
- European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Heidelberg, Germany
| | - Lynn E Lambert
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Adam F Sander
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Sophia Hundt
- European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Heidelberg, Germany
| | - Sachy Orr-Gonzalez
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christoph Mikkel Janitzek
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Alicia J Spiegel
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Marvin L Thomas
- Division of Veterinary Resources, Office of Research Services, National Institutes of Health, Bethesda, MD, USA
| | - Martha C Nason
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Maryonne Snow-Smith
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Emma K Barnafo
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joseph Shiloach
- Biotechnology Unit, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Beth B Chen
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Steven Nadakal
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kendrick Highsmith
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tarik Ouahes
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Solomon Conteh
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ankur Sharma
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Holly Torano
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Brandi Butler
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Karine Reiter
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kelly M Rausch
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Puthupparampil V Scaria
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Charles Anderson
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David L Narum
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ali Salanti
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Michal Fried
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Thor G Theander
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Benoit Gamain
- Université Paris Cité and Université des Antilles, INSERM, BIGR, Paris, France
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
11
|
Sagara I, Healy SA, Assadou MH, Kone M, Swihart BJ, Kwan JL, Fintzi J, Sissoko K, Kamate B, Samake Y, Guindo MA, Doucoure M, Niaré K, Dolo A, Diarra B, Rausch KM, Narum DL, Jones DS, MacDonald NJ, Zhu D, Gorres JP, Imeru A, Mohan R, Thera I, Zaidi I, Salazar-Miralles F, Duan J, Neal J, Morrison RD, Muratova O, Sylla D, O'Connell EM, Wu Y, Hume JCC, Coulibaly MB, Anderson CF, Traore SF, Doumbo OK, Duffy PE. Malaria transmission-blocking vaccines Pfs230D1-EPA and Pfs25-EPA in Alhydrogel in healthy Malian adults; a phase 1, randomised, controlled trial. THE LANCET. INFECTIOUS DISEASES 2023; 23:1266-1279. [PMID: 37499679 PMCID: PMC10615700 DOI: 10.1016/s1473-3099(23)00276-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Malaria transmission-blocking vaccines target mosquito-stage parasites and will support elimination programmes. Gamete vaccine Pfs230D1-EPA/Alhydrogel induced superior activity to zygote vaccine Pfs25-EPA/Alhydrogel in malaria-naive US adults. Here, we compared these vaccines in malaria-experienced Malians. METHODS We did a pilot safety study then double-blind, block-randomised, comparator-controlled main-phase trial in malaria-intense Bancoumana, Mali. 18-50-year-old healthy non-pregnant, non-breastfeeding consenting adult residents were randomly assigned (1:1:1:1) to receive four doses at months 0, 1, 4·5, and 16·5 of either 47 μg Pfs25, 40 μg Pfs230D1 or comparator (Twinrix or Menactra)-all co-administered with normal saline for blinding-or 47 μg Pfs25 plus 40 μg Pfs230D1 co-administered. We documented safety and tolerability (primary endpoint in the as-treated populations) and immunogenicity (secondary endpoint in the as-treated populations: ELISA, standard-membrane-feeding assay, and mosquito direct skin feed assay). This trial is registered at ClinicalTrials.gov, NCT02334462. FINDINGS Between March 19, and June 2, 2015, we screened 471 individuals. Of 225 enrolled for the pilot and main cohorts, we randomly assigned 25 participants to pilot safety cohort groups of five (20%) to receive a two-dose series of Pfs25-EPA/Alhydrogel (16 μg), Pfs230D1-EPA/Alhydrogel (15 μg) or comparator, followed by Pfs25-EPA/Alhydrogel (16 μg) plus Pfs230D1-EPA/Alhydrogel (15 μg) or comparator plus saline. For the main cohort, we enrolled 200 participants between May 11 and June 2, 2015, to receive a four-dose series of 47 μg Pfs25-EPA/Alhydrogel plus saline (n=50 [25%]; Pfs25), 40 μg Pfs230D1-EPA/Alhydrogel plus saline (n=49 [25%]; Pfs230D1), 47 μg Pfs25-EPA/Alhydrogel plus 40 μg Pfs230D1-EPA/Alhydrogel (n=50 [25%]; Pfs25 plus Pfs230D1), or comparator (Twinrix or Menactra) plus saline (n=51 [25%]). Vaccinations were well tolerated in the pilot safety and main phases. Most vaccinees became seropositive after two Pfs230D1 or three Pfs25 doses; peak titres increased with each dose thereafter (Pfs230D1 geometric mean: 77·8 [95% CI 56·9-106·3], 146·4 [108·3-198·0], and 410·2 [301·6-558·0]; Pfs25 geometric mean 177·7 [130·3-242·4] and 315·7 [209·9-474·6]). Functional activity (mean peak transmission-reducing activity) appeared for Pfs230D1 (74·5% [66·6-82·5]) and Pfs25 plus Pfs230D1 (68·6% [57·3-79·8]), after the third dose and after the fourth dose (88·9% [81·7-96·2] for Pfs230D1 and 85·0% [78·4-91·5] Pfs25 plus Pfs230D1) but not for Pfs25 (58·2% [49·1-67·3] after the third dose and 58·2% [48·5-67·9] after the fourth dose). Pfs230D1 transmission-reducing activity (73·7% [64·1-83·3]) persisted 10 weeks after the fourth dose. Transmission-reducing activity of 80% was estimated at 1659 ELISA units for Pfs25, 218 for Pfs230D1, and 223 for Pfs230D1 plus Pfs25. After 3850 direct skin feed assays, 35 participants (12 Pfs25, eight Pfs230D1, five Pfs25 plus Pfs230D1, and ten comparator) had transmitted parasites at least once. The proportion of positive assays in vaccine groups (Pfs25 33 [3%] of 982 [-0·013 to 0·014], Pfs230D1 22 [2%] of 954 [-0·005 to 0·027], and combination 11 [1%] of 940 [-0·024 to 0·002]) did not differ from that of the comparator (22 [2%] of 974), nor did Pfs230D1 and combination groups differ (-0·024 to 0·001). INTERPRETATION Pfs230D1 but not Pfs25 vaccine induces durable serum functional activity in Malian adults. Direct skin feed assays detect parasite transmission to mosquitoes but increased event rates are needed to assess vaccine effectiveness. FUNDING Intramural Research Program of the National Institute of Allergy and Infectious Diseases and US National Institutes of Health.
Collapse
Affiliation(s)
- 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, Mali
| | - Sara A Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Mahamadoun H Assadou
- 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, Mali
| | - Mamady Kone
- 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, Mali
| | - Bruce J Swihart
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer L Kwan
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan Fintzi
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kourane Sissoko
- 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, Mali
| | - Bourama Kamate
- 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, Mali
| | - Yacouba Samake
- 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, Mali
| | - Merepen A Guindo
- 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, Mali
| | - M'Bouye Doucoure
- 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, Mali
| | - Karamoko Niaré
- 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, Mali
| | - Amagana Dolo
- 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, Mali
| | - Balla Diarra
- 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, Mali
| | - Kelly M Rausch
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - David L Narum
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - David S Jones
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Nicholas J MacDonald
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Daming Zhu
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - J Patrick Gorres
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Alemush Imeru
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Rathy Mohan
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Ismaila Thera
- 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, Mali
| | - Irfan Zaidi
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Fernando Salazar-Miralles
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Junhui Duan
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Jillian Neal
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Robert D Morrison
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Olga Muratova
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Daman Sylla
- 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, Mali
| | - Elise M O'Connell
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yimin Wu
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Jen C C Hume
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Mamadou B Coulibaly
- 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, Mali
| | - Charles F Anderson
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Sekou F Traore
- 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, Mali
| | - Ogobara K Doumbo
- 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, Mali
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
12
|
Abstract
Malaria is a mosquito-borne disease caused by protozoan parasites of the genus Plasmodium. Despite significant declines in malaria-attributable morbidity and mortality over the last two decades, it remains a major public health burden in many countries. This underscores the critical need for improved strategies to prevent, treat and control malaria if we are to ultimately progress towards the eradication of this disease. Ideally, this will include the development and deployment of a highly effective malaria vaccine that is able to induce long-lasting protective immunity. There are many malaria vaccine candidates in development, with more than a dozen of these in clinical development. RTS,S/AS01 (also known as Mosquirix) is the most advanced malaria vaccine and was shown to have modest efficacy against clinical malaria in phase III trials in 5- to 17-month-old infants. Following pilot implementation trials, the World Health Organisation has recommended it for use in Africa in young children who are most at risk of infection with P. falciparum, the deadliest of the human malaria parasites. It is well recognised that more effective malaria vaccines are needed. In this review, we discuss malaria vaccine candidates that have progressed into clinical evaluation and highlight the most advanced candidates: Sanaria's irradiated sporozoite vaccine (PfSPZ Vaccine), the chemoattenuated sporozoite vaccine (PfSPZ-CVac), RTS,S/AS01 and the novel malaria vaccine candidate, R21, which displayed promising, high-level efficacy in a recent small phase IIb trial in Africa.
Collapse
Affiliation(s)
- Danielle I Stanisic
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia.
| | - Michael F Good
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia.
| |
Collapse
|
13
|
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
|
14
|
Rausch KM, Barnafo EK, Lambert LE, Muratova O, Gorres JP, Anderson C, Narum DL, Wu Y, Morrison RD, Zaidi I, Duffy PE. Preclinical evaluations of Pfs25-EPA and Pfs230D1-EPA in AS01 for a vaccine to reduce malaria transmission. iScience 2023; 26:107192. [PMID: 37485364 PMCID: PMC10359932 DOI: 10.1016/j.isci.2023.107192] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 02/15/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
Malaria transmission-blocking vaccine candidates Pfs25-EPA and Pfs230D1-EPA target sexual stage development of Plasmodium falciparum parasites in the mosquito host, thereby reducing mosquito infectivity. When formulated on Alhydrogel, Pfs25-EPA has demonstrated safety and immunogenicity in a phase 1 field trial, while Pfs230D1-EPA has shown superior activity to Pfs25-EPA in a phase 1 US trial and has entered phase 2 field trials. Development continues to enhance immunogenicity of these candidates toward producing a vaccine to reduce malaria transmission (VRMT) with both pre-erythrocytic (i.e., anti-infection) and transmission-blocking components. GSK Adjuvant Systems have demonstrated successful potency in pre-erythrocytic vaccine trials and might offer a common platform for VRMT development. Here, we describe preclinical evaluations of Pfs25-EPA and Pfs230D1-EPA nanoparticles with GSK platforms. Formulations were stable after a series of assessments and induced superior antibody titers and functional activity in CD-1 mice, compared to Alhydrogel formulations of the same antigens.
Collapse
Affiliation(s)
- Kelly M. Rausch
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Emma K. Barnafo
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lynn E. Lambert
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Olga Muratova
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - J. Patrick Gorres
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Charles Anderson
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David L. Narum
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yimin Wu
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert D. Morrison
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Irfan Zaidi
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patrick E. Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
15
|
MacDonald NJ, Singh K, Reiter K, Nguyen V, Shimp R, Gittis AG, Chen B, Burkhardt M, Zhang B, Wang Z, Herrera R, Moler M, Lee DY, Orr-Gonzalez S, Herrod J, Lambert LE, Rausch KM, Muratova O, Jones DS, Wu Y, Jin AJ, Garboczi DN, Duffy PE, Narum DL. Structural and immunological differences in Plasmodium falciparum sexual stage transmission-blocking vaccines comprised of Pfs25-EPA nanoparticles. NPJ Vaccines 2023; 8:56. [PMID: 37061547 PMCID: PMC10105769 DOI: 10.1038/s41541-023-00655-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 03/29/2023] [Indexed: 04/17/2023] Open
Abstract
Development of a malaria vaccine that blocks transmission of different parasite stages to humans and mosquitoes is considered critical for elimination efforts. A vaccine using Pfs25, a protein on the surface of zygotes and ookinetes, is under investigation as a transmission-blocking vaccine (TBV) that would interrupt parasite passage from mosquitoes to humans. The most extensively studied Pfs25 TBVs use Pichia pastoris-produced recombinant forms of Pfs25, chemically conjugated to a recombinant carrier protein, ExoProtein A (EPA). The recombinant form of Pfs25 first used in humans was identified as Pfs25H, which contained a total of 14 heterologous amino acid residues located at the amino- and carboxyl-termini including a His6 affinity tag. A second recombinant Pfs25, identified as Pfs25M, was produced to remove the heterologous amino acid residues and conjugated to EPA (Pfs25M-EPA). Here, monomeric Pfs25M was characterized biochemically and biophysically for identity, purity, and integrity including protein structure to assess its comparability with Pfs25H. Although the biological activities of Pfs25H and Pfs25M, whether generated by monomeric forms or conjugated nanoparticles, appeared similar, fine-mapping studies with two transmission-blocking monoclonal antibodies detected structural and immunological differences. In addition, evaluation of antisera generated against conjugated Pfs25H or Pfs25M nanoparticles in nonhuman primates identified polyclonal IgG that recognized these structural differences.
Collapse
Affiliation(s)
- Nicholas J MacDonald
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Kavita Singh
- Structural Biology Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Karine Reiter
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Vu Nguyen
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Richard Shimp
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Apostolos G Gittis
- Structural Biology Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Beth Chen
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Martin Burkhardt
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Baoshan Zhang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Zhixiong Wang
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Raul Herrera
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Mackenzie Moler
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Duck-Yeon Lee
- National Heart, Lung, and Blood Institute, Bethesda, MD, 20814, USA
| | - Sachy Orr-Gonzalez
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Jessica Herrod
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Lynn E Lambert
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Kelly M Rausch
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Olga Muratova
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - David S Jones
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Yimin Wu
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Albert J Jin
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - David N Garboczi
- Structural Biology Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA
| | - David L Narum
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 29 Lincoln Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
16
|
Tang WK, Coelho CH, Miura K, Nguemwo Tentokam BC, Salinas ND, Narum DL, Healy SA, Sagara I, Long CA, Duffy PE, Tolia NH. A human antibody epitope map of Pfs230D1 derived from analysis of individuals vaccinated with a malaria transmission-blocking vaccine. Immunity 2023; 56:433-443.e5. [PMID: 36792576 PMCID: PMC9989938 DOI: 10.1016/j.immuni.2023.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/07/2022] [Accepted: 01/13/2023] [Indexed: 02/16/2023]
Abstract
Pfs230 domain 1 (Pfs230D1) is an advanced malaria transmission-blocking vaccine antigen demonstrating high functional activity in clinical trials. However, the structural and functional correlates of transmission-blocking activity are not defined. Here, we characterized a panel of human monoclonal antibodies (hmAbs) elicited in vaccinees immunized with Pfs230D1. These hmAbs exhibited diverse transmission-reducing activity, yet all bound to Pfs230D1 with nanomolar affinity. We compiled epitope-binning data for seventeen hmAbs and structures of nine hmAbs complexes to construct a high-resolution epitope map and revealed that potent transmission-reducing hmAbs bound to one face of Pfs230D1, while non-potent hmAbs bound to the opposing side. The structure of Pfs230D1D2 revealed that non-potent transmission-reducing epitopes were occluded by the second domain. The hmAb epitope map delineated binary hmAb combinations that synergized for extremely high-potency, transmission-reducing activity. This work provides a high-resolution guide for structure-based design of enhanced immunogens and informs diagnostics that measure the transmission-reducing response.
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
| | - 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, MD, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Bergeline C Nguemwo Tentokam
- Pathogenesis and Immunity 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
| | - David L Narum
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, 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, MD, USA
| | - Issaka Sagara
- Malaria Research and Training Center, University of Sciences, Techniques, and Technology, Bamako, Mali
| | - Carole A Long
- 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, MD, USA; Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 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
|
17
|
Mariano RMDS, Gonçalves AAM, de Oliveira DS, Ribeiro HS, Pereira DFS, Santos IS, Lair DF, da Silva AV, Galdino AS, Chávez-Fumagalli MA, da Silveira-Lemos D, Dutra WO, Giunchetti RC. A Review of Major Patents on Potential Malaria Vaccine Targets. Pathogens 2023; 12:pathogens12020247. [PMID: 36839519 PMCID: PMC9959516 DOI: 10.3390/pathogens12020247] [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: 12/31/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Malaria is a parasitic infection that is a great public health concern and is responsible for high mortality rates worldwide. Different strategies have been employed to improve disease control, demonstrating the ineffectiveness of controlling vectors, and parasite resistance to antimalarial drugs requires the development of an effective preventive vaccine. There are countless challenges to the development of such a vaccine directly related to the parasite's complex life cycle. After more than four decades of basic research and clinical trials, the World Health Organization (WHO) has recommended the pre-erythrocytic Plasmodium falciparum (RTS, S) malaria vaccine for widespread use among children living in malaria-endemic areas. However, there is a consensus that major improvements are needed to develop a vaccine with a greater epidemiological impact in endemic areas. This review discusses novel strategies for malaria vaccine design taking the target stages within the parasite cycle into account. The design of the multi-component vaccine shows considerable potential, especially as it involves transmission-blocking vaccines (TBVs) that eliminate the parasite's replication towards sporozoite stage parasites during a blood meal of female anopheline mosquitoes. Significant improvements have been made but additional efforts to achieve an efficient vaccine are required to improve control measures. Different strategies have been employed, thus demonstrating the ineffectiveness in controlling vectors, and parasite resistance to antimalarial drugs requires the development of a preventive vaccine. Despite having a vaccine in an advanced stage of development, such as the RTS, S malaria vaccine, the search for an effective vaccine against malaria is far from over. This review discusses novel strategies for malaria vaccine design taking into account the target stages within the parasite's life cycle.
Collapse
Affiliation(s)
- Reysla Maria da Silveira Mariano
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Ana Alice Maia Gonçalves
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Diana Souza de Oliveira
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Helen Silva Ribeiro
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Diogo Fonseca Soares Pereira
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Ingrid Soares Santos
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Daniel Ferreira Lair
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Augusto Ventura da Silva
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Alexsandro Sobreira Galdino
- Laboratory of Biotechnology of Microorganisms, Federal University of São João Del-Rei, Divinópolis CEP 35501-296, MG, Brazil
| | - Miguel Angel Chávez-Fumagalli
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Urb. San José S/N, Arequipa 04000, Peru
| | - Denise da Silveira-Lemos
- Campus Jaraguá, University José of Rosário Vellano, UNIFENAS, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Walderez Ornelas Dutra
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Rodolfo Cordeiro Giunchetti
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
- Correspondence: or ; Tel.: +55-31-3409-3003
| |
Collapse
|
18
|
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
|
19
|
Duffy PE. Current approaches to malaria vaccines. Curr Opin Microbiol 2022; 70:102227. [PMID: 36343566 PMCID: PMC11127243 DOI: 10.1016/j.mib.2022.102227] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/27/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022]
Abstract
The complex Plasmodium life cycle offers different vaccine approaches with distinct parasitological and clinical effects. The approaches and their rationales were established decades ago: vaccines targeting pre-erythrocytic (sporozoite and liver-stage) parasites prevent infection, those to blood-stage parasites reduce disease, and those to sexual-stage parasites or mosquito vector reduce transmission and eliminate malaria through herd immunity. The pre-erythrocytic RTS,S vaccine (Mosquirix, GlaskoSmithKline (GSK)), recommended by WHO in 2021, reduces clinical malaria in children. Knowledge of parasite biology, host-parasite interactions, and immune mechanisms is informing new concepts to improve on RTS,S and to target other parasite stages. This review emphasizes vaccine approaches and candidates currently in the clinic or likely to enter clinical testing soon.
Collapse
Affiliation(s)
- Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
20
|
Structural analysis of Plasmodium falciparum ookinete surface antigen Pfs28 relevant for malaria vaccine design. Sci Rep 2022; 12:19556. [PMID: 36379968 PMCID: PMC9664031 DOI: 10.1038/s41598-022-24054-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Pfs28 is a Plasmodium falciparum malaria transmission-blocking vaccine candidate that is anchored to the parasite surface through a C-terminal glycosylphosphatidylinositol (GPI) moiety, and plays a role in parasite survival in the mosquito midgut. Pfs28 contains epidermal growth factor (EGF)-like domains and is part of a family of sexual stage malaria proteins that includes the related vaccine antigen Pfs25. The lack of structural definition of Pfs28 and the immune response to this candidate has limited further malaria vaccine development for this antigen. Here, we present the crystal structure of Pfs28, examine its conservation with P. vivax Pvs28, and evaluate the cross-reactivity of Pfs28 to antibodies that recognize Pfs25. Pfs28 is comprised of four EGF-like domains stabilized by ten disulfide bridges with an overall architecture that highly resembles Pfs25. Despite the high sequence and structural similarity between these antigens, no cross reactivity of Pfs28 to anti-Pfs25 monoclonal antibodies could be demonstrated.
Collapse
|
21
|
Coelho CH, Galson JD, Trück J, Duffy PE. B cell clonal expansion and mutation in the immunoglobulin heavy chain variable domain in response to Pfs230 and Pfs25 malaria vaccines. Int J Parasitol 2022; 52:707-710. [PMID: 34896314 PMCID: PMC9177897 DOI: 10.1016/j.ijpara.2021.11.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 12/26/2022]
Abstract
Malaria transmission-blocking vaccines induce antibodies that target Plasmodium in the mosquito vector. We recently reported that Pfs230 vaccine achieves activity superior to Pfs25 in humans. Here, we describe clonal expansion in the variable region of immunoglobulin heavy chains (VH) of antigen-specific single B cells collected from humans immunised with Pfs230D1-EPA or Pfs25-EPA conjugate vaccines formulated in Alhydrogel®. Based on studies of CD27+ memory B cells following Pfs230 vaccination, clonal expansion and somatic hypermutation was seen in four of five subjects. Pfs25 did not induce sufficient CD27+ cells for sorting; based instead on CD19+ Pfs25-reactive B cells, clonal expansion was only seen in two of five subjects. Clonal expansions and mutations in Pfs230-specific single B cells combined with the enhanced activity of Pfs230 antibodies by complement, might justify the outstanding activity of Pfs230D1 as a TBV candidate.
Collapse
Affiliation(s)
- Camila H Coelho
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Jacob D Galson
- Division of Immunology, University Children's Hospital Zurich and Clinical Research Center, University of Zurich, Switzerland; Alchemab Therapeutics Ltd, London, United Kingdom
| | - Johannes Trück
- Division of Immunology, University Children's Hospital Zurich and Clinical Research Center, University of Zurich, Switzerland
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA.
| |
Collapse
|
22
|
Iyori M, Blagborough AM, Mizuno T, Abe YI, Nagaoka M, Hori N, Yamagoshi I, Da DF, Gregory WF, Hasyim AA, Yamamoto Y, Sakamoto A, Yoshida K, Mizukami H, Shida H, Yoshida S. Sterile protection and transmission blockade by a multistage anti-malarial vaccine in the pre-clinical study. Front Immunol 2022; 13:1005476. [PMID: 36248835 PMCID: PMC9558734 DOI: 10.3389/fimmu.2022.1005476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
The Malaria Vaccine Technology Roadmap 2013 (World Health Organization) aims to develop safe and effective vaccines by 2030 that will offer at least 75% protective efficacy against clinical malaria and reduce parasite transmission. Here, we demonstrate a highly effective multistage vaccine against both the pre-erythrocytic and sexual stages of Plasmodium falciparum that protects and reduces transmission in a murine model. The vaccine is based on a viral-vectored vaccine platform, comprising a highly-attenuated vaccinia virus strain, LC16m8Δ (m8Δ), a genetically stable variant of a licensed and highly effective Japanese smallpox vaccine LC16m8, and an adeno-associated virus (AAV), a viral vector for human gene therapy. The genes encoding P. falciparum circumsporozoite protein (PfCSP) and the ookinete protein P25 (Pfs25) are expressed as a Pfs25-PfCSP fusion protein, and the heterologous m8Δ-prime/AAV-boost immunization regimen in mice provided both 100% protection against PfCSP-transgenic P. berghei sporozoites and up to 100% transmission blocking efficacy, as determined by a direct membrane feeding assay using parasites from P. falciparum-positive, naturally-infected donors from endemic settings. Remarkably, the persistence of vaccine-induced immune responses were over 7 months and additionally provided complete protection against repeated parasite challenge in a murine model. We propose that application of the m8Δ/AAV malaria multistage vaccine platform has the potential to contribute to the landmark goals of the malaria vaccine technology roadmap, to achieve life-long sterile protection and high-level transmission blocking efficacy.
Collapse
Affiliation(s)
- Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | | | - Tetsushi Mizuno
- Department of Parasitology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Yu-ichi Abe
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | - Mio Nagaoka
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | - Naoto Hori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | - Iroha Yamagoshi
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | - Dari F. Da
- Département de Biologie Médicale et Santé Publique, Unité Paludisme et Maladies Tropicales Négligées, Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - William F. Gregory
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Ammar A. Hasyim
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | - Yutaro Yamamoto
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | - Akihiko Sakamoto
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | - Kunitaka Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| | - Hiroaki Mizukami
- Division of Gene Therapy, Jichi Medical University, Tochigi, Japan
| | - Hisatoshi Shida
- Institute for Genetic Medicine, Hokkaido University, Hokkaido, Japan
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Ishikawa, Japan
| |
Collapse
|
23
|
Hasyim AA, Iyori M, Mizuno T, Abe YI, Yamagoshi I, Yusuf Y, Syafira I, Sakamoto A, Yamamoto Y, Mizukami H, Shida H, Yoshida S. Adeno-associated virus-based malaria booster vaccine following attenuated replication-competent vaccinia virus LC16m8Δ priming. Parasitol Int 2022; 92:102652. [PMID: 36007703 DOI: 10.1016/j.parint.2022.102652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022]
Abstract
We previously demonstrated that boosting with adeno-associated virus (AAV) type 1 (AAV1) can induce highly effective and long-lasting protective immune responses against malaria parasites when combined with replication-deficient adenovirus priming in a rodent model. In the present study, we compared the efficacy of two different AAV serotypes, AAV1 and AAV5, as malaria booster vaccines following priming with the attenuated replication-competent vaccinia virus strain LC16m8Δ (m8Δ), which harbors the fusion gene encoding both the pre-erythrocytic stage protein, Plasmodium falciparum circumsporozoite (PfCSP) and the sexual stage protein (Pfs25) in a two-dose heterologous prime-boost immunization regimen. Both regimens, m8Δ/AAV1 and m8Δ/AAV5, induced robust anti-PfCSP and anti-Pfs25 antibodies. To evaluate the protective efficacy, the mice were challenged with sporozoites twice after immunization. At the first sporozoite challenge, m8Δ/AAV5 achieved 100% sterile protection whereas m8Δ/AAV1 achieved 70% protection. However, at the second challenge, 100% of the surviving mice from the first challenge were protected in the m8Δ/AAV1 group whereas only 55.6% of those in the m8Δ/AAV5 group were protected. Regarding the transmission-blocking efficacy, we found that both immunization regimens induced high levels of transmission-reducing activity (>99%) and transmission-blocking activity (>95%). Our data indicate that the AAV5-based multistage malaria vaccine is as effective as the AAV1-based vaccine when administered following an m8Δ-based vaccine. These results suggest that AAV5 could be a viable alternate vaccine vector as a malaria booster vaccine.
Collapse
Affiliation(s)
- Ammar A Hasyim
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Ishikawa 920-1192, Japan
| | - Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Ishikawa 920-1192, Japan
| | - Tetsushi Mizuno
- Department of Global Infectious Diseases, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa 920-0934, Japan
| | - Yu-Ichi Abe
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Ishikawa 920-1192, Japan
| | - Iroha Yamagoshi
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Ishikawa 920-1192, Japan
| | - Yenni Yusuf
- Department of Parasitology, Faculty of Medicine, Hasanuddin University, Makassar, Sulawesi Selatan 90245, Indonesia
| | - Intan Syafira
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Ishikawa 920-1192, Japan
| | - Akihiko Sakamoto
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Ishikawa 920-1192, Japan
| | - Yutaro Yamamoto
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Ishikawa 920-1192, Japan
| | - Hiroaki Mizukami
- Division of Gene Therapy, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Hisatoshi Shida
- Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido 060-0815, Japan
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Ishikawa 920-1192, Japan.
| |
Collapse
|
24
|
Scaria PV, Rowe CG, Chen BB, Dickey TH, Renn JP, Lambert LE, Barnafo EK, Rausch KM, Tolia NH, Duffy PE. Protein-protein conjugation enhances the immunogenicity of SARS-CoV-2 receptor-binding domain (RBD) vaccines. iScience 2022; 25:104739. [PMID: 35846379 PMCID: PMC9270177 DOI: 10.1016/j.isci.2022.104739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 06/06/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022] Open
Abstract
Several effective SARS-CoV-2 vaccines have been developed using different technologies. Although these vaccines target the isolates collected early in the pandemic, many have protected against serious illness from newer variants. Nevertheless, efficacy has diminished against successive variants and the need for effective and affordable vaccines persists especially in the developing world. Here, we adapted our protein-protein conjugate vaccine technology to generate a vaccine based on receptor-binding domain (RBD) antigen. RBD was conjugated to a carrier protein, EcoCRM®, to generate two types of conjugates: crosslinked and radial conjugates. In the crosslinked conjugate, antigen and carrier are chemically crosslinked; in the radial conjugate, the antigen is conjugated to the carrier by site-specific conjugation. With AS01 adjuvant, both conjugates showed enhanced immunogenicity in mice compared to RBD, with a Th1 bias. In hACE2 binding inhibition and pseudovirus neutralization assays, sera from mice vaccinated with the radial conjugate demonstrated strong functional activity.
Collapse
Affiliation(s)
- Puthupparampil V. Scaria
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903, USA
| | - Chris G. Rowe
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903, USA
| | - Beth B. Chen
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903, USA
| | - Thayne H. Dickey
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903, USA
| | - Jonathan P. Renn
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903, USA
| | - Lynn E. Lambert
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903, USA
| | - Emma K. Barnafo
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903, USA
| | - Kelly M. Rausch
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903, USA
| | - Niraj H. Tolia
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903, USA
| | - Patrick E. Duffy
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903, USA
| |
Collapse
|
25
|
Jagannath DK, Valiyaparambil A, Viswanath VK, Hurakadli MA, Kamariah N, Jafer AC, Patole C, Pradhan S, Kumar N, Lakshminarasimhan A. Refolding and characterization of a diabody against Pfs25, a vaccine candidate of Plasmodium falciparum. Anal Biochem 2022; 655:114830. [DOI: 10.1016/j.ab.2022.114830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/28/2022] [Accepted: 07/20/2022] [Indexed: 11/28/2022]
|
26
|
Duffy PE. The Virtues and Vices of Pfs230: From Vaccine Concept to Vaccine Candidate. Am J Trop Med Hyg 2022; 107:tpmd211337. [PMID: 35895391 DOI: 10.4269/ajtmh.21-1337] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 02/23/2022] [Indexed: 02/18/2024] Open
Abstract
Among the Plasmodium falciparum surface antigens reported by Richard Carter and his colleagues decades ago, Pfs230 is currently the target of the most advanced candidate for a malaria transmission-blocking vaccine. First identified by its orthologue in the avian malaria parasite Plasmodium gallinaceum, the large cysteine-rich 14-domain Pfs230 antigen is displayed on the surface of gametes that emerge in the mosquito midgut. Gametes lacking Pfs230 cannot bind to red blood cells nor develop further into oocysts. Human antibodies against Pfs230 lyse gametes in the presence of complement, which largely explains serum transmission-blocking activity in Pfs230 antisera. A protein-protein conjugate vaccine that incorporates the first domain of the Pfs230 antigen induced greater serum transmission-reducing activity versus a similarly manufactured Pfs25 vaccine in U.S. trials, and is currently in phase II field trials in Mali.
Collapse
|
27
|
Kaslow DC. Efforts to Develop Pfs25 Vaccines. Am J Trop Med Hyg 2022; 107:tpmd211326. [PMID: 35895392 DOI: 10.4269/ajtmh.21-1326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/10/2022] [Indexed: 02/18/2024] Open
Abstract
Acknowledging the fallibilities of recalling events from more than three decades ago, the recollection of Richard Carter's impact on the identification and development of Pfs25, a major surface protein of Plasmodium falciparum zygotes and ookinetes, and target of malaria transmission-blocking vaccines, remains unassailable. In fondest memories of Richard Carter's many contributions, herein retells some memorable events along the tortuous journey toward the development of Pfs25 vaccines.
Collapse
|
28
|
de Jong RM, Singh SK, Teelen K, van de Vegte-Bolmer M, van Gemert GJ, Stone WJR, Locke E, Plieskatt J, Theisen M, Bousema T, Jore MM. Heterologous Expression and Evaluation of Novel Plasmodium falciparum Transmission Blocking Vaccine Candidates. Front Immunol 2022; 13:909060. [PMID: 35812379 PMCID: PMC9259988 DOI: 10.3389/fimmu.2022.909060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/17/2022] [Indexed: 11/27/2022] Open
Abstract
Malaria transmission blocking vaccines (TBV) aim to induce antibodies that can interrupt Plasmodium falciparum development in the mosquito midgut and thereby prevent onward malaria transmission. A limited number of TBV candidates have been identified and only three (Pfs25, Pfs230 and Pfs48/45) have entered clinical testing. While one of these candidates may emerge as a highly potent TBV candidate, it is premature to determine if they will generate sufficiently potent and sustained responses. It is therefore important to explore novel candidate antigens. We recently analyzed sera from naturally exposed individuals and found that the presence and/or intensity of antibodies against 12 novel putative surface expressed gametocyte antigens was associated with transmission reducing activity. In this study, protein fragments of these novel TBV candidates were designed and heterologously expressed in Drosophila melanogaster S2 cells and Lactococcus lactis. Eleven protein fragments, covering seven TBV candidates, were successfully produced. All tested antigens were recognized by antibodies from individuals living in malaria-endemic areas, indicating that native epitopes are present. All antigens induced antigen-specific antibody responses in mice. Two antigens induced antibodies that recognized a native protein in gametocyte extract, and antibodies elicited by four antigens recognized whole gametocytes. In particular, we found that antigen Pf3D7_0305300, a putative transporter, is abundantly expressed on the surface of gametocytes. However, none of the seven novel TBV candidates expressed here induced an antibody response that reduced parasite development in the mosquito midgut as assessed in the standard membrane feeding assay. Altogether, the antigen fragments used in this study did not prove to be promising transmission blocking vaccine constructs, but led to the identification of two gametocyte surface proteins that may provide new leads for studying gametocyte biology.
Collapse
Affiliation(s)
- Roos M. de Jong
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Susheel K. Singh
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Karina Teelen
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Will J. R. Stone
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Emily Locke
- PATH‘s Malaria Vaccine Initiative, Washington, DC, United States
| | - Jordan Plieskatt
- PATH‘s Malaria Vaccine Initiative, Washington, DC, United States
| | - Michael Theisen
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Teun Bousema
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Matthijs M. Jore
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
- *Correspondence: Matthijs M. Jore,
| |
Collapse
|
29
|
Mulamba C, Williams C, Kreppel K, Ouedraogo JB, Olotu AI. Evaluation of the Pfs25-IMX313/Matrix-M malaria transmission-blocking candidate vaccine in endemic settings. Malar J 2022; 21:159. [PMID: 35655174 PMCID: PMC9161629 DOI: 10.1186/s12936-022-04173-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/02/2022] [Indexed: 11/10/2022] Open
Abstract
Malaria control relies heavily on the use of anti-malarial drugs and insecticides against malaria parasites and mosquito vectors. Drug and insecticide resistance threatens the effectiveness of conventional malarial interventions; alternative control approaches are, therefore, needed. The development of malaria transmission-blocking vaccines that target the sexual stages in humans or mosquito vectors is among new approaches being pursued. Here, the immunological mechanisms underlying malaria transmission blocking, status of Pfs25-based vaccines are viewed, as well as approaches and capacity for first in-human evaluation of a transmission-blocking candidate vaccine Pfs25-IMX313/Matrix-M administered to semi-immune healthy individuals in endemic settings. It is concluded that institutions in low and middle income settings should be supported to conduct first-in human vaccine trials in order to stimulate innovative research and reduce the overdependence on developed countries for research and local interventions against many diseases of public health importance.
Collapse
Affiliation(s)
- Charles Mulamba
- Interventions & Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania.,Nelson Mandela African Institution of Science and Technology, Tengeru, P. O. Box 447, Arusha, Tanzania
| | - Chris Williams
- The Jenner Institute, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7DQ, UK
| | - Katharina Kreppel
- Nelson Mandela African Institution of Science and Technology, Tengeru, P. O. Box 447, Arusha, Tanzania
| | | | - Ally I Olotu
- Interventions & Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania.
| |
Collapse
|
30
|
Wang PP, Jiang X, Bai J, Yang F, Yu X, Wu Y, Zheng W, Zhang Y, Cui L, Liu F, Zhu X, Cao Y. Characterization of PSOP26 as an ookinete surface antigen with improved transmission-blocking activity when fused with PSOP25. Parasit Vectors 2022; 15:175. [PMID: 35606790 PMCID: PMC9125894 DOI: 10.1186/s13071-022-05294-8] [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] [Received: 02/12/2022] [Accepted: 04/18/2022] [Indexed: 11/26/2022] Open
Abstract
Background The Plasmodium zygote-to-ookinete developmental transition is an essential step for establishing an infection in the mosquito vector, and antigens expressed during this stage are potential targets for transmission-blocking vaccines (TBVs). The secreted ookinete protein 26 (PSOP26) is a newly identified ookinete surface protein. The anti-PSOP26 serum has moderate transmission-blocking activity, indicating the benefit of further investigating this protein as a target for TBVs. Methods The function of psop26 was analyzed by targeted gene disruption. A chimeric PSOP25-PSOP26 protein was expressed in the Escherichia coli system. The PSOP25-PSOP26 fusion protein, along with mixed (PSOP25 + PSOP26) or single proteins (PSOP26 or PSOP25), were used for the immunization of mice. The antibody titers and immunogenicity of individual sera were analyzed by enzyme-linked immunoassay (ELISA), indirect immunofluorescence assay (IFA), and Western blot. The transmission-blocking activity of sera from different immunization schemes was assessed using in vitro and in vivo assays. Results PSOP26 is a surface protein expressed in Plasmodium gametes and ookinetes. The protein is dispensable for asexual blood-stage development, gametogenesis, and zygote formation, but is essential for the zygote-to-ookinete developmental transition. Specifically, both the prevalence of infections and oocyst densities were decreased in mosquitoes fed on psop26-null mutants. Mixtures of individual PSOP25 and PSOP26 fragments (PSOP25 + PSOP26), as well as chimeras (PSOP25-PSOP26), elicited high antibody levels in mice, with no immunological interference. Antisera against the mixed and fusion proteins elicited higher transmission-reducing activity (TRA) than antisera against the single PSOP26 antigen, but comparable to antisera against PSOP25 antigen alone. Conclusions PSOP26 plays a critical role in the zygote-to-ookinete developmental transition. PSOP25 is a promising TBV candidate that could be used alone to target the ookinete stage. Supplementary Information The online version contains supplementary material available at 10.1186/s13071-022-05294-8.
Collapse
Affiliation(s)
- Peng-Peng Wang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China.,Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xuefeng Jiang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Jie Bai
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Fan Yang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Xinxin Yu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yudi Wu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Wenqi Zheng
- Department of Clinical Laboratory, Affiliated Hospital of Inner Mongolian Medical University, Inner Mongolia, Huhhot, 150000, China
| | - Yongzhe Zhang
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China.,Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Suite 304, Tampa, FL, 33612-9415, USA
| | - Fei Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Xiaotong Zhu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
31
|
Ochwedo KO, Onyango SA, Omondi CJ, Orondo PW, Ondeto BM, Lee MC, Atieli HE, Ogolla SO, Githeko AK, Otieno ACA, Mukabana WR, Yan G, Zhong D, Kazura JW. Signatures of selection and drivers for novel mutation on transmission-blocking vaccine candidate Pfs25 gene in western Kenya. PLoS One 2022; 17:e0266394. [PMID: 35390042 PMCID: PMC8989228 DOI: 10.1371/journal.pone.0266394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 03/20/2022] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Leading transmission-blocking vaccine candidates such as Plasmodium falciparum surface protein 25 (Pfs25 gene) may undergo antigenic alterations which may render them ineffective or allele-specific. This study examines the level of genetic diversity, signature of selection and drivers of Pfs25 polymorphisms of parasites population in regions of western Kenya with varying malaria transmission intensities. METHODS Dry blood spots (DBS) were collected in 2018 and 2019 from febrile outpatients with malaria at health facilities in malaria-endemic areas of Homa Bay, Kisumu (Chulaimbo) and the epidemic-prone highland area of Kisii. Parasites DNA were extracted from DBS using Chelex method. Species identification was performed using real-time PCR. The 460 base pairs (domains 1-4) of the Pfs25 were amplified and sequenced for a total of 180 P. falciparum-infected blood samples. RESULTS Nine of ten polymorphic sites were identified for the first time. Overall, Pfs25 exhibited low nucleotide diversity (0.04×10-2) and low mutation frequencies (1.3% to 7.7%). Chulaimbo had the highest frequency (15.4%) of mutated sites followed by Kisii (6.7%) and Homa Bay (5.1%). Neutrality tests of Pfs25 variations showed significant negative values of Tajima's D (-2.15, p<0.01) and Fu's F (-10.91, p<0.001) statistics tests. Three loci pairs (123, 372), (364, 428) and (390, 394) were detected to be under linkage disequilibrium and none had history of recombination. These results suggested that purifying selection and inbreeding might be the drivers of the observed variation in Pfs25. CONCLUSION Given the low level of nucleotide diversity, it is unlikely that a Pfs25 antigen-based vaccine would be affected by antigenic variations. However, continued monitoring of Pfs25 immunogenic domain 3 for possible variants that might impact vaccine antibody binding is warranted.
Collapse
Affiliation(s)
- Kevin O. Ochwedo
- Department of Biology, Faculty of Science and Technology, University of Nairobi, Nairobi, Kenya
- Sub-Saharan Africa International Centre for Excellence in Malaria Research, Homa Bay, Kenya
| | - Shirley A. Onyango
- Sub-Saharan Africa International Centre for Excellence in Malaria Research, Homa Bay, Kenya
- School of Zoological Sciences, Kenyatta University, Nairobi, Kenya
| | - Collince J. Omondi
- Department of Biology, Faculty of Science and Technology, University of Nairobi, Nairobi, Kenya
- Sub-Saharan Africa International Centre for Excellence in Malaria Research, Homa Bay, Kenya
| | - Pauline W. Orondo
- Sub-Saharan Africa International Centre for Excellence in Malaria Research, Homa Bay, Kenya
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Benyl M. Ondeto
- Department of Biology, Faculty of Science and Technology, University of Nairobi, Nairobi, Kenya
- Sub-Saharan Africa International Centre for Excellence in Malaria Research, Homa Bay, Kenya
| | - Ming-Chieh Lee
- Program in Public Health, College of Health Sciences, University of California, Irvine, Irvine, California, United States of America
| | - Harrysone E. Atieli
- Sub-Saharan Africa International Centre for Excellence in Malaria Research, Homa Bay, Kenya
- School of Public Health and Community Development, Maseno University, Kisumu, Kenya
| | - Sidney O. Ogolla
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Andrew K. Githeko
- Sub-Saharan Africa International Centre for Excellence in Malaria Research, Homa Bay, Kenya
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Antony C. A. Otieno
- Department of Biology, Faculty of Science and Technology, University of Nairobi, Nairobi, Kenya
| | - Wolfgang R. Mukabana
- Department of Biology, Faculty of Science and Technology, University of Nairobi, Nairobi, Kenya
- Sub-Saharan Africa International Centre for Excellence in Malaria Research, Homa Bay, Kenya
| | - Guiyun Yan
- Program in Public Health, College of Health Sciences, University of California, Irvine, Irvine, California, United States of America
| | - Daibin Zhong
- Program in Public Health, College of Health Sciences, University of California, Irvine, Irvine, California, United States of America
| | - James W. Kazura
- Centre for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, United States of America
| |
Collapse
|
32
|
Arévalo-Herrera M, Miura K, Solano E, Ramírez JS, Long CA, Corradin G, Herrera S. Immunogenicity of full-length P. vivax rPvs48/45 protein formulations in BALB/c mice. Vaccine 2022; 40:133-140. [PMID: 34802791 PMCID: PMC9109962 DOI: 10.1016/j.vaccine.2021.11.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/12/2021] [Accepted: 11/11/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND Pvs48/45 is a Plasmodium vivax gametocyte surface protein involved in the parasite fertilization process. Previous studies showed that Pvs48/45 proteins expressed in Escherichia coli (E. coli) and Chinese hamster ovary (CHO) cells were highly immunoreactive with sera from malaria-endemic areas and highly immunogenic in animal models. Here the immunogenicity in mice of three different vaccine formulations was compared. METHODS Recombinant (r) Pvs48/45 proteins were expressed in E. coli and CHO, purified, formulated in Alhydrogel, GLA-SE and Montanide ISA-51 adjuvants and used to immunize BALB/c mice. Animals were immunized on days 0, 20 and 40, and serum samples were collected for serological analyses of specific antibody responses using ELISA and immunofluorescence (IFAT). Additionally, ex-vivo transmission-reducing activity (TRA) of sera on P. vivax gametocyte-infected human blood fed to Anopheles albimanus in direct membrane feeding assays (DMFA) was evaluated. RESULTS Most immunized animals seroconverted after the first immunization, and some developed antibody peaks of 106 with all adjuvants. However, the three adjuvant formulations induced different antibody responses and TRA efficacy. While GLA-SE formulations of both proteins induced similar antibody profiles, Montanide ISA-51 formulations resulted in higher and longer-lasting antibody titers with CHO-rPvs48/45 than with the E. coli formulation. Although the CHO protein formulated in Alhydrogel generated a high initial antibody peak, antibody responses to both proteins rapidly waned. Likewise, anti-Pvs48/45 antibodies displayed differential recognition of the parasite proteins in IFAT and ex vivo blockade of parasite transmission to mosquitoes. The CHO-rPvs48/45 formulated in Montanide ISA-51 induced the most effective ex vivo parasite blockage. CONCLUSIONS Three out of six vaccine formulations elicited antibodies with ex vivo TRA. The CHO-rPvs48/45 Montanide ISA-51 formulation induced the most stable antibody response, recognizing the native protein and the most robust ex vivo TRA. These results encourage further testing of the vaccine potential of this protein.
Collapse
Affiliation(s)
- Myriam Arévalo-Herrera
- Malaria Vaccine and Drug Development Center, Cali, Colombia.,Caucaseco Scientific Research Center, Cali, Colombia.,Corresponding author: Myriam Arevalo-Herrera, PhD, , Phone: (+57 2) 521 6232, Fax: (+57 2) 521 6228, Malaria Vaccine and Drug Development Center, Carrera 37 2 Bis # 5E-08. Cali, Colombia
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, 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
| | | | - Sócrates Herrera
- Malaria Vaccine and Drug Development Center, Cali, Colombia.,Caucaseco Scientific Research Center, Cali, Colombia
| |
Collapse
|
33
|
Takashima E, Tachibana M, Morita M, Nagaoka H, Kanoi BN, Tsuboi T. Identification of Novel Malaria Transmission-Blocking Vaccine Candidates. Front Cell Infect Microbiol 2021; 11:805482. [PMID: 34917521 PMCID: PMC8670312 DOI: 10.3389/fcimb.2021.805482] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 11/16/2021] [Indexed: 01/02/2023] Open
Abstract
Control measures have significantly reduced malaria morbidity and mortality in the last two decades; however, the downward trends have stalled and have become complicated by the emergence of COVID-19. Significant efforts have been made to develop malaria vaccines, but currently only the RTS,S/AS01 vaccine against Plasmodium falciparum has been recommended by the WHO, for widespread use among children in sub-Saharan Africa. The efficacy of RTS,S/AS01 is modest, and therefore the development of more efficacious vaccines is still needed. In addition, the development of transmission-blocking vaccines (TBVs) to reduce the parasite transmission from humans to mosquitoes is required toward the goal of malaria elimination. Few TBVs have reached clinical development, and challenges include low immunogenicity or high reactogenicity in humans. Therefore, novel approaches to accelerate TBV research and development are urgently needed, especially novel TBV candidate discovery. In this mini review we summarize the progress in TBV research and development, novel TBV candidate discovery, and discuss how to accelerate novel TBV candidate discovery.
Collapse
Affiliation(s)
- Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Mayumi Tachibana
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Japan
| | - Masayuki Morita
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Bernard N Kanoi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Takafumi Tsuboi
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama, Japan
| |
Collapse
|
34
|
Scaria PV, Anderson C, Muratova O, Alani N, Trinh HV, Nadakal ST, Zaidi I, Lambert L, Beck Z, Barnafo EK, Rausch KM, Rowe C, Chen B, Matyas GR, Rao M, Alving CR, Narum DL, Duffy PE. Malaria transmission-blocking conjugate vaccine in ALFQ adjuvant induces durable functional immune responses in rhesus macaques. NPJ Vaccines 2021; 6:148. [PMID: 34887448 PMCID: PMC8660773 DOI: 10.1038/s41541-021-00407-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/03/2021] [Indexed: 12/14/2022] Open
Abstract
Malaria transmission-blocking vaccines candidates based on Pfs25 and Pfs230 have advanced to clinical studies. Exoprotein A (EPA) conjugate of Pfs25 in Alhydrogel® developed functional immunity in humans, with limited durability. Pfs230 conjugated to EPA (Pfs230D1-EPA) with liposomal adjuvant AS01 is currently in clinical trials in Mali. Studies with these conjugates revealed that non-human primates are better than mice to recapitulate the human immunogenicity and functional activity. Here, we evaluated the effect of ALFQ, a liposomal adjuvant consisting of TLR4 agonist and QS21, on the immunogenicity of Pfs25-EPA and Pfs230D1-EPA in Rhesus macaques. Both conjugates generated strong antibody responses and functional activity after two vaccinations though activity declined rapidly. A third vaccination of Pfs230D1-EPA induced functional activity lasting at least 9 months. Antibody avidity increased with each vaccination and correlated strongly with functional activity. IgG subclass analysis showed induction of Th1 and Th2 subclass antibody levels that correlated with activity.
Collapse
Affiliation(s)
- Puthupparampil V. Scaria
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Charles Anderson
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Olga Muratova
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Nada Alani
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Hung V. Trinh
- grid.507680.c0000 0001 2230 3166U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910 USA ,grid.201075.10000 0004 0614 9826Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817 USA
| | - Steven T. Nadakal
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Irfan Zaidi
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Lynn Lambert
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Zoltan Beck
- grid.507680.c0000 0001 2230 3166U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910 USA ,grid.201075.10000 0004 0614 9826Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817 USA ,grid.410513.20000 0000 8800 7493Present Address: Pfizer, Vaccine Research and Development, Pearl River, NY USA
| | - Emma K. Barnafo
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Kelly M. Rausch
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Chris Rowe
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Beth Chen
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Gary R. Matyas
- grid.507680.c0000 0001 2230 3166U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910 USA
| | - Mangala Rao
- grid.507680.c0000 0001 2230 3166U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910 USA
| | - Carl R. Alving
- grid.507680.c0000 0001 2230 3166U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910 USA
| | - David L. Narum
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Patrick E. Duffy
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| |
Collapse
|
35
|
Tachibana M, Takashima E, Morita M, Sattabongkot J, Ishino T, Culleton R, Torii M, Tsuboi T. Plasmodium vivax transmission-blocking vaccines: Progress, challenges and innovation. Parasitol Int 2021; 87:102525. [PMID: 34896614 DOI: 10.1016/j.parint.2021.102525] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 12/04/2021] [Indexed: 12/12/2022]
Abstract
Existing control measures have significantly reduced malaria morbidity and mortality in the last two decades, although these reductions are now stalling. Significant efforts have been undertaken to develop malaria vaccines. Recently, extensive progress in malaria vaccine development has been made for Plasmodium falciparum. To date, only the RTS,S/AS01 vaccine has been tested in Phase 3 clinical trials and is now under implementation, despite modest efficacy. Therefore, the development of a malaria transmission-blocking vaccine (TBV) will be essential for malaria elimination. Only a limited number of TBVs have reached pre-clinical or clinical development with several major challenges impeding their development, including low immunogenicity in humans. TBV development efforts against P. vivax, the second major cause of malaria morbidity, lag far behind those for P. falciparum. In this review we summarize the latest progress, challenges and innovations in P. vivax TBV research and discuss how to accelerate its development.
Collapse
Affiliation(s)
- Mayumi Tachibana
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan.
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan.
| | - Masayuki Morita
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan.
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand.
| | - Tomoko Ishino
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan.
| | - Richard Culleton
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan.
| | - Motomi Torii
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan; Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan.
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan.
| |
Collapse
|
36
|
Keleta Y, Ramelow J, Cui L, Li J. Molecular interactions between parasite and mosquito during midgut invasion as targets to block malaria transmission. NPJ Vaccines 2021; 6:140. [PMID: 34845210 PMCID: PMC8630063 DOI: 10.1038/s41541-021-00401-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 11/01/2021] [Indexed: 11/21/2022] Open
Abstract
Despite considerable effort, malaria remains a major public health burden. Malaria is caused by five Plasmodium species and is transmitted to humans via the female Anopheles mosquito. The development of malaria vaccines against the liver and blood stages has been challenging. Therefore, malaria elimination strategies advocate integrated measures, including transmission-blocking approaches. Designing an effective transmission-blocking strategy relies on a sophisticated understanding of the molecular mechanisms governing the interactions between the mosquito midgut molecules and the malaria parasite. Here we review recent advances in the biology of malaria transmission, focusing on molecular interactions between Plasmodium and Anopheles mosquito midgut proteins. We provide an overview of parasite and mosquito proteins that are either targets for drugs currently in clinical trials or candidates of promising transmission-blocking vaccines.
Collapse
Affiliation(s)
- Yacob Keleta
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA
| | - Julian Ramelow
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Liwang Cui
- College of Public Health, University of South Florida, Tampa, FL, 33612, USA
| | - Jun Li
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA.
- Biomolecular Science Institute, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
37
|
Sookpongthai P, Utayopas K, Sitthiyotha T, Pengsakul T, Kaewthamasorn M, Wangkanont K, Harnyuttanakorn P, Chunsrivirot S, Pattaradilokrat S. Global diversity of the gene encoding the Pfs25 protein-a Plasmodium falciparum transmission-blocking vaccine candidate. Parasit Vectors 2021; 14:571. [PMID: 34749796 PMCID: PMC8574928 DOI: 10.1186/s13071-021-05078-6] [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] [Received: 08/03/2021] [Accepted: 10/21/2021] [Indexed: 11/29/2022] Open
Abstract
Background Vaccines against the sexual stages of the malarial parasite Plasmodium falciparum are indispensable for controlling malaria and abrogating the spread of drug-resistant parasites. Pfs25, a surface antigen of the sexual stage of P. falciparum, is a leading candidate for transmission-blocking vaccine development. While clinical trials have reported that Pfs25-based vaccines are safe and effective in inducing transmission-blocking antibodies, the extent of the genetic diversity of Pfs25 in malaria endemic populations has rarely been studied. Thus, this study aimed to investigate the global diversity of Pfs25 in P. falciparum populations. Methods A database of 307 Pfs25 sequences of P. falciparum was established. Population genetic analyses were performed to evaluate haplotype and nucleotide diversity, analyze haplotypic distribution patterns of Pfs25 in different geographical populations, and construct a haplotype network. Neutrality tests were conducted to determine evidence of natural selection. Homology models of the Pfs25 haplotypes were constructed, subjected to molecular dynamics (MD), and analyzed in terms of flexibility and percentages of secondary structures. Results The Pfs25 gene of P. falciparum was found to have 11 unique haplotypes. Of these, haplotype 1 (H1) and H2, the major haplotypes, represented 70% and 22% of the population, respectively, and were dominant in Asia, whereas only H1 was dominant in Africa, Central America, and South America. Other haplotypes were rare and region-specific, resulting in unique distribution patterns in different geographical populations. The diversity in Pfs25 originated from ten single-nucleotide polymorphism (SNP) loci located in the epidermal growth factor (EGF)-like domains and anchor domain. Of these, an SNP at position 392 (GGA/GCA), resulting in amino acid substitution 131 (Gly/Ala), defined the two major haplotypes. The MD results showed that the structures of H1 and H2 variants were relatively similar. Limited polymorphism in Pfs25 could likely be due to negative selection. Conclusions The study successfully established a Pfs25 sequence database that can become an essential tool for monitoring vaccine efficacy, designing assays for detecting malaria carriers, and conducting epidemiological studies of P. falciparum. The discovery of the two major haplotypes, H1 and H2, and their conserved structures suggests that the current Pfs25-based vaccines could be used globally for malaria control. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-021-05078-6.
Collapse
Affiliation(s)
- Pornpawee Sookpongthai
- M.Sc. program in Zoology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Korawich Utayopas
- Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thassanai Sitthiyotha
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Theerakamol Pengsakul
- Faculty of Medical Technology, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Morakot Kaewthamasorn
- Veterinary Parasitology Research Unit, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kittikhun Wangkanont
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | | | - Surasak Chunsrivirot
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand.,Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | | |
Collapse
|
38
|
Zaric M, Marini A, Nielsen CM, Gupta G, Mekhaiel D, Pham TP, Elias SC, Taylor IJ, de Graaf H, Payne RO, Li Y, Silk SE, Williams C, Hill AVS, Long CA, Miura K, Biswas S. Poor CD4 + T Cell Immunogenicity Limits Humoral Immunity to P. falciparum Transmission-Blocking Candidate Pfs25 in Humans. Front Immunol 2021; 12:732667. [PMID: 34659219 PMCID: PMC8515144 DOI: 10.3389/fimmu.2021.732667] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/07/2021] [Indexed: 11/13/2022] Open
Abstract
Plasmodium falciparum transmission-blocking vaccines (TBVs) targeting the Pfs25 antigen have shown promise in mice but the same efficacy has never been achieved in humans. We have previously published pre-clinical data related to a TBV candidate Pfs25-IMX313 encoded in viral vectors which was very promising and hence progressed to human clinical trials. The results from the clinical trial of this vaccine were very modest. Here we unravel why, contrary to mice, this vaccine has failed to induce robust antibody (Ab) titres in humans to elicit transmission-blocking activity. We examined Pfs25-specific B cell and T follicular helper (Tfh) cell responses in mice and humans after vaccination with Pfs25-IMX313 encoded by replication-deficient chimpanzee adenovirus serotype 63 (ChAd63) and the attenuated orthopoxvirus modified vaccinia virus Ankara (MVA) delivered in the heterologous prime-boost regimen via intramuscular route. We found that after vaccination, the Pfs25-IMX313 was immunologically suboptimal in humans compared to mice in terms of serum Ab production and antigen-specific B, CD4+ and Tfh cell responses. We identified that the key determinant for the poor anti-Pfs25 Ab formation in humans was the lack of CD4+ T cell recognition of Pfs25-IMX313 derived peptide epitopes. This is supported by correlations established between the ratio of proliferated antigen-specific CD4+/Tfh-like T cells, CXCL13 sera levels, and the corresponding numbers of circulating Pfs25-specific memory B cells, that consequently reflected on antigen-specific IgG sera levels. These correlations can inform the design of next-generation Pfs25-based vaccines for robust and durable blocking of malaria transmission.
Collapse
Affiliation(s)
- Marija Zaric
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Arianna Marini
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Carolyn M Nielsen
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Gaurav Gupta
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - David Mekhaiel
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Thao P Pham
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD, United States
| | - Sean C Elias
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Iona J Taylor
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Hans de Graaf
- NIHR Clinical Research Facility, University Hospital Southampton NHS Foundation Trust and Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ruth O Payne
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Yuanyuan Li
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Sarah E Silk
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Chris Williams
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Adrian V S Hill
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD, United States
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD, United States
| | - Sumi Biswas
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
39
|
de Jong RM, Meerstein-Kessel L, Da DF, Nsango S, Challenger JD, van de Vegte-Bolmer M, van Gemert GJ, Duarte E, Teyssier N, Sauerwein RW, Churcher TS, Dabire RK, Morlais I, Locke E, Huynen MA, Bousema T, Jore MM. Monoclonal antibodies block transmission of genetically diverse Plasmodium falciparum strains to mosquitoes. NPJ Vaccines 2021; 6:101. [PMID: 34385463 PMCID: PMC8361195 DOI: 10.1038/s41541-021-00366-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/29/2021] [Indexed: 11/09/2022] Open
Abstract
Malaria parasite transmission to mosquitoes relies on the uptake of sexual stage parasites during a blood meal and subsequent formation of oocysts on the mosquito midgut wall. Transmission-blocking vaccines (TBVs) and monoclonal antibodies (mAbs) target sexual stage antigens to interrupt human-to-mosquito transmission and may form important tools for malaria elimination. Although most epitopes of these antigens are considered highly conserved, little is known about the impact of natural genetic diversity on the functional activity of transmission-blocking antibodies. Here we measured the efficacy of three mAbs against leading TBV candidates (Pfs48/45, Pfs25 and Pfs230) in transmission assays with parasites from naturally infected donors compared to their efficacy against the strain they were raised against (NF54). Transmission-reducing activity (TRA) was measured as reduction in mean oocyst intensity. mAb 45.1 (α-Pfs48/45) and mAb 4B7 (α-Pfs25) reduced transmission of field parasites from almost all donors with IC80 values similar to NF54. Sequencing of oocysts that survived high mAb concentrations did not suggest enrichment of escape genotypes. mAb 2A2 (α-Pfs230) only reduced transmission of parasites from a minority of the donors, suggesting that it targets a non-conserved epitope. Using six laboratory-adapted strains, we revealed that mutations in one Pfs230 domain correlate with mAb gamete surface binding and functional TRA. Our findings demonstrate that, despite the conserved nature of sexual stage antigens, minor sequence variation can significantly impact the efficacy of transmission-blocking mAbs. Since mAb 45.1 shows high potency against genetically diverse strains, our findings support its further clinical development and may inform Pfs48/45 vaccine design.
Collapse
Affiliation(s)
- Roos M de Jong
- Department of Medical Microbiology and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lisette Meerstein-Kessel
- Department of Medical Microbiology and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Center for Molecular and Biomolecular Informatics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dari F Da
- Institut de Recherche en Sciences de la Santé, Direction Régionale, Bobo Dioulasso, Burkina Faso
| | - Sandrine Nsango
- Malaria Research Laboratory, OCEAC, Yaoundé, Cameroon
- Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala, Cameroon
| | - Joseph D Challenger
- Medical Research Council Centre for Global Infections Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Marga van de Vegte-Bolmer
- Department of Medical Microbiology and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Geert-Jan van Gemert
- Department of Medical Microbiology and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Elias Duarte
- EPPIcenter Research Program, Division of HIV, ID, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Noam Teyssier
- EPPIcenter Research Program, Division of HIV, ID, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Robert W Sauerwein
- Department of Medical Microbiology and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- TropIQ Health Sciences, Nijmegen, Netherlands
| | - Thomas S Churcher
- Medical Research Council Centre for Global Infections Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Roch K Dabire
- Institut de Recherche en Sciences de la Santé, Direction Régionale, Bobo Dioulasso, Burkina Faso
| | - Isabelle Morlais
- Malaria Research Laboratory, OCEAC, Yaoundé, Cameroon
- MIVEGEC, Université Montpellier, IRD, CNRS, Montpellier, France
| | - Emily Locke
- PATH's Malaria Vaccine Initiative, Washington, DC, USA
| | - Martijn A Huynen
- Center for Molecular and Biomolecular Informatics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Teun Bousema
- Department of Medical Microbiology and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Matthijs M Jore
- Department of Medical Microbiology and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
40
|
de Graaf H, Payne RO, Taylor I, Miura K, Long CA, Elias SC, Zaric M, Minassian AM, Silk SE, Li L, Poulton ID, Baker M, Draper SJ, Gbesemete D, Brendish NJ, Martins F, Marini A, Mekhaiel D, Edwards NJ, Roberts R, Vekemans J, Moyle S, Faust SN, Berrie E, Lawrie AM, Hill F, Hill AVS, Biswas S. Safety and Immunogenicity of ChAd63/MVA Pfs25-IMX313 in a Phase I First-in-Human Trial. Front Immunol 2021; 12:694759. [PMID: 34335606 PMCID: PMC8318801 DOI: 10.3389/fimmu.2021.694759] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/29/2021] [Indexed: 11/13/2022] Open
Abstract
Background Transmission blocking vaccines targeting the sexual-stages of the malaria parasite could play a major role to achieve elimination and eradication of malaria. The Plasmodium falciparum Pfs25 protein (Pfs25) is the most clinically advanced candidate sexual-stage antigen. IMX313, a complement inhibitor C4b-binding protein that forms heptamers with the antigen fused to it, improve antibody responses. This is the first time that viral vectors have been used to induce antibodies in humans against an antigen that is expressed only in the mosquito vector. Methods Clinical trial looking at safety and immunogenicity of two recombinant viral vectored vaccines encoding Pfs25-IMX313 in healthy malaria-naive adults. Replication-deficient chimpanzee adenovirus serotype 63 (ChAd63) and the attenuated orthopoxvirus modified vaccinia virus Ankara (MVA), encoding Pfs25-IMX313, were delivered by the intramuscular route in a heterologous prime-boost regimen using an 8-week interval. Safety data and samples for immunogenicity assays were taken at various time-points. Results The reactogenicity of the vaccines was similar to that seen in previous trials using the same viral vectors encoding other antigens. The vaccines were immunogenic and induced both antibody and T cell responses against Pfs25, but significant transmission reducing activity (TRA) was not observed in most volunteers by standard membrane feeding assay. Conclusion Both vaccines were well tolerated and demonstrated a favorable safety profile in malaria-naive adults. However, the transmission reducing activity of the antibodies generated were weak, suggesting the need for an alternative vaccine formulation. Trial Registration Clinicaltrials.gov NCT02532049.
Collapse
Affiliation(s)
- Hans de Graaf
- NIHR Clinical Research Facility, University Hospital Southampton NHS Foundation Trust and Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ruth O Payne
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Iona Taylor
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Carol A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Sean C Elias
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Marija Zaric
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Sarah E Silk
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Lee Li
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Ian D Poulton
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Megan Baker
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Diane Gbesemete
- NIHR Clinical Research Facility, University Hospital Southampton NHS Foundation Trust and Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Nathan J Brendish
- NIHR Clinical Research Facility, University Hospital Southampton NHS Foundation Trust and Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Filipa Martins
- NIHR Clinical Research Facility, University Hospital Southampton NHS Foundation Trust and Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Arianna Marini
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - David Mekhaiel
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Nick J Edwards
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Rachel Roberts
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Sarah Moyle
- Clinical Biomanufacturing Facility, University of Oxford, Oxford, United Kingdom
| | - Saul N Faust
- NIHR Clinical Research Facility, University Hospital Southampton NHS Foundation Trust and Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Eleanor Berrie
- Clinical Biomanufacturing Facility, University of Oxford, Oxford, United Kingdom
| | - Alison M Lawrie
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Adrian V S Hill
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Sumi Biswas
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
41
|
Arévalo-Herrera M, Miura K, Cespedes N, Echeverry C, Solano E, Castellanos A, Ramirez JS, Miranda A, Kajava AV, Long C, Corradin G, Herrera S. Immunoreactivity of Sera From Low to Moderate Malaria-Endemic Areas Against Plasmodium vivax r Pvs48/45 Proteins Produced in Escherichia coli and Chinese Hamster Ovary Systems. Front Immunol 2021; 12:634738. [PMID: 34248932 PMCID: PMC8264144 DOI: 10.3389/fimmu.2021.634738] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 05/25/2021] [Indexed: 11/13/2022] Open
Abstract
P48/45 is a conserved gametocyte antigen involved in Plasmodium parasite fertilization. A recombinant Plasmodium vivax P48/45 (Pvs48/45) protein expressed in Escherichia coli (E. coli) was highly antigenic and immunogenic in experimental animals and elicited specific transmission-blocking (TB) antibodies in a previous pilot study. Here, a similar Pvs48/45 gene was expressed in Chinese Hamster Ovary (CHO) cells and we compared its immunoreactivity with the E. coli product. Specific antibody titers were determined using plasma from Colombian individuals (n=227) living in endemic areas where both P. vivax and P. falciparum are prevalent and from Guatemala (n=54) where P. vivax is highly prevalent. In Colombia, plasma seroprevalence to CHO-rPvs48/45 protein was 46.3%, while for E. coli-rPvs48/45 protein was 36.1% (p<0.001). In Guatemala, the sero prevalence was 24.1% and 14.8% (p<0.001), respectively. Reactivity index (RI) against both proteins showed an age-dependent increase. IgG2 was the predominant subclass and the antibody avidity index evaluated by ELISA ranged between 4-6 mol/L. Ex vivo P. vivax mosquito direct membrane feeding assays (DMFA) performed in presence of study plasmas, displayed significant parasite transmission-blocking (TB), however, there was no direct correlation between antibody titers and oocysts transmission reduction activity (%TRA). Nevertheless, DMFA with CHO rPvs48/45 affinity purified IgG showed a dose response; 90.2% TRA at 100 μg/mL and 71.8% inhibition at 10 μg/mL. In conclusion, the CHO-rPvs48/45 protein was more immunoreactive in most of the malaria endemic places studied, and CHO-rPvs48/45 specific IgG showed functional activity, supporting further testing of the protein vaccine potential.
Collapse
Affiliation(s)
- Myriam Arévalo-Herrera
- Immunology Department, Malaria Vaccine and Drug Development Center, Cali, Colombia
- Immunology Department, Caucaseco Scientific Research Center, Cali, Colombia
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Nora Cespedes
- Immunology Department, Malaria Vaccine and Drug Development Center, Cali, Colombia
| | - Carlos Echeverry
- Immunology Department, Malaria Vaccine and Drug Development Center, Cali, Colombia
| | - Eduardo Solano
- Immunology Department, Caucaseco Scientific Research Center, Cali, Colombia
| | - Angélica Castellanos
- Immunology Department, Malaria Vaccine and Drug Development Center, Cali, Colombia
| | | | - Adolfo Miranda
- Parasitology Department, Centro Nacional de Epidemiología (CNE), Guatemala City, Guatemala
| | - Andrey V. Kajava
- Centre de Recherche en Biologie Cellulaire de Montpellier, Université Montpellier, Montpellier, France
| | - Carole Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | | | - Sócrates Herrera
- Immunology Department, Malaria Vaccine and Drug Development Center, Cali, Colombia
- Immunology Department, Caucaseco Scientific Research Center, Cali, Colombia
| |
Collapse
|
42
|
Tripathi AK, Oakley MS, Verma N, Mlambo G, Zheng H, Meredith SM, Essuman E, Puri A, Skelton RA, Takeda K, Majam V, Quakyi IA, Locke E, Morin M, Miura K, Long CA, Kumar S. Plasmodium falciparum Pf77 and male development gene 1 as vaccine antigens that induce potent transmission-reducing antibodies. Sci Transl Med 2021; 13:eabg2112. [PMID: 34108248 PMCID: PMC11018285 DOI: 10.1126/scitranslmed.abg2112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 04/22/2021] [Indexed: 12/14/2022]
Abstract
Malaria vaccines that disrupt the Plasmodium life cycle in mosquitoes and reduce parasite transmission in endemic areas are termed transmission-blocking vaccines (TBVs). Despite decades of research, there are only a few Plasmodium falciparum antigens that indisputably and reproducibly demonstrate transmission-blocking immunity. So far, only two TBV candidates have advanced to phase 1/2 clinical testing with limited success. By applying an unbiased transcriptomics-based approach, we have identified Pf77 and male development gene 1 (PfMDV-1) as two P. falciparum TBV antigens that, upon immunization, induced antibodies that caused reductions in oocyst counts in Anopheles mosquito midguts in a standard membrane feeding assay. In-depth studies were performed to characterize the genetic diversity of, stage-specific expression by, and natural immunity to these two molecules to evaluate their suitability as TBV candidates. Pf77 and PfMDV-1 display limited antigenic polymorphism, are pan-developmentally expressed within the parasite, and induce naturally occurring antibodies in Ghanaian adults, which raises the prospect of natural boosting of vaccine-induced immune response in endemic regions. Together, these biological properties suggest that Pf77 and PfMDV-1 may warrant further investigation as TBV candidates.
Collapse
Affiliation(s)
- Abhai K Tripathi
- Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Miranda S Oakley
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Nitin Verma
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Godfree Mlambo
- Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Hong Zheng
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Scott M Meredith
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Edward Essuman
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Ankit Puri
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Richard A Skelton
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Kazuyo Takeda
- Division of Bacterial, Parasitic, and Allergenic Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Victoria Majam
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | | | - Emily Locke
- PATH-Malaria Vaccine Initiative, Washington, DC 20001, USA
| | | | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, 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
| | - Sanjai Kumar
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA.
| |
Collapse
|
43
|
Nguyen B, Tolia NH. Protein-based antigen presentation platforms for nanoparticle vaccines. NPJ Vaccines 2021; 6:70. [PMID: 33986287 PMCID: PMC8119681 DOI: 10.1038/s41541-021-00330-7] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/22/2021] [Indexed: 02/08/2023] Open
Abstract
Modern vaccine design has sought a minimalization approach, moving to the isolation of antigens from pathogens that invoke a strong neutralizing immune response. This approach has created safer vaccines but may limit vaccine efficacy due to poor immunogenicity. To combat global diseases such as COVID-19, malaria, and AIDS there is a clear urgency for more effective next-generation vaccines. One approach to improve the immunogenicity of vaccines is the use of nanoparticle platforms that present a repetitive array of antigen on its surface. This technology has been shown to improve antigen presenting cell uptake, lymph node trafficking, and B-cell activation through increased avidity and particle size. With a focus on design, we summarize natural platforms, methods of antigen attachment, and advancements in generating self-assembly that have led to new engineered platforms. We further examine critical parameters that will direct the usage and development of more effective platforms.
Collapse
Affiliation(s)
- Brian Nguyen
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
| | - Niraj H Tolia
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA.
| |
Collapse
|
44
|
Soon MSF, Nalubega M, Boyle MJ. T-follicular helper cells in malaria infection and roles in antibody induction. OXFORD OPEN IMMUNOLOGY 2021; 2:iqab008. [PMID: 36845571 PMCID: PMC9914587 DOI: 10.1093/oxfimm/iqab008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 01/29/2023] Open
Abstract
Immunity to malaria is mediated by antibodies that block parasite replication to limit parasite burden and prevent disease. Cytophilic antibodies have been consistently shown to be associated with protection, and recent work has improved our understanding of the direct and Fc-mediated mechanisms of protective antibodies. Antibodies also have important roles in vaccine-mediated immunity. Antibody induction is driven by the specialized CD4+ T cells, T-follicular helper (Tfh) cells, which function within the germinal centre to drive B-cell activation and antibody induction. In humans, circulating Tfh cells can be identified in peripheral blood and are differentiated into subsets that appear to have pathogen/vaccination-specific roles in antibody induction. Tfh cell responses are essential for protective immunity from Plasmodium infection in murine models of malaria. Our understanding of the activation of Tfh cells during human malaria infection and the importance of different Tfh cell subsets in antibody development is still emerging. This review will discuss our current knowledge of Tfh cell activation and development in malaria, and the potential avenues and pitfalls of targeting Tfh cells to improve malaria vaccines.
Collapse
Affiliation(s)
- Megan S F Soon
- Department of Infectious Diseases, QIMR-Berghofer, 300 Herston Road, Herston, QLD, 4006, Australia
| | - Mayimuna Nalubega
- Infectious Diseases Research Collaboration, Tororo District Hospital, Tororo, Uganda
| | - Michelle J Boyle
- Department of Infectious Diseases, QIMR-Berghofer, 300 Herston Road, Herston, QLD, 4006, Australia,Correspondence address. QIMR Berghofer Medical Research Institute, Brisbane, Australia. E-mail:
| |
Collapse
|
45
|
Healy SA, Anderson C, Swihart BJ, Mwakingwe A, Gabriel EE, Decederfelt H, Hobbs CV, Rausch KM, Zhu D, Muratova O, Herrera R, Scaria PV, MacDonald NJ, Lambert LE, Zaidi I, Coelho CH, Renn JP, Wu Y, Narum DL, Duffy PE. Pfs230 yields higher malaria transmission-blocking vaccine activity than Pfs25 in humans but not mice. J Clin Invest 2021; 131:146221. [PMID: 33561016 PMCID: PMC8011888 DOI: 10.1172/jci146221] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/03/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUNDVaccines that block human-to-mosquito Plasmodium transmission are needed for malaria eradication, and clinical trials have targeted zygote antigen Pfs25 for decades. We reported that a Pfs25 protein-protein conjugate vaccine formulated in alum adjuvant induced serum functional activity in both US and Malian adults. However, antibody levels declined rapidly, and transmission-reducing activity required 4 vaccine doses. Functional immunogenicity and durability must be improved before advancing transmission-blocking vaccines further in clinical development. We hypothesized that the prefertilization protein Pfs230 alone or in combination with Pfs25 would improve functional activity.METHODSTransmission-blocking vaccine candidates based on gamete antigen Pfs230 or Pfs25 were conjugated with Exoprotein A, formulated in Alhydrogel, and administered to mice, rhesus macaques, and humans. Antibody levels were measured by ELISA and transmission-reducing activity was assessed by the standard membrane feeding assay.RESULTSPfs25-EPA/Alhydrogel and Pfs230D1-EPA/Alhydrogel induced similar serum functional activity in mice, but Pfs230D1-EPA induced significantly greater activity in rhesus monkeys that was enhanced by complement. In US adults, 2 vaccine doses induced complement-dependent activity in 4 of 5 Pfs230D1-EPA/Alhydrogel recipients but no significant activity in 5 Pfs25-EPA recipients, and combination with Pfs25-EPA did not increase activity over Pfs230D1-EPA alone.CONCLUSIONThe complement-dependent functional immunogenicity of Pfs230D1-EPA represents a significant improvement over Pfs25-EPA in this comparative study. The rhesus model is more predictive of the functional human immune response to Pfs230D1 than is the mouse model.TRIAL REGISTRATIONClinicalTrials.gov NCT02334462.FUNDINGIntramural Research Program of the National Institute of Allergy and Infectious Diseases, National Institutes of Health.
Collapse
Affiliation(s)
- Sara A Healy
- Laboratory of Malaria Immunology and Vaccinology, and
| | | | - Bruce J Swihart
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Erin E Gabriel
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.,Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Hope Decederfelt
- Pharmacy Department, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - Daming Zhu
- Laboratory of Malaria Immunology and Vaccinology, and
| | - Olga Muratova
- Laboratory of Malaria Immunology and Vaccinology, and
| | - Raul Herrera
- Laboratory of Malaria Immunology and Vaccinology, and
| | | | | | | | - Irfan Zaidi
- Laboratory of Malaria Immunology and Vaccinology, and
| | | | | | - Yimin Wu
- Laboratory of Malaria Immunology and Vaccinology, and
| | - David L Narum
- Laboratory of Malaria Immunology and Vaccinology, and
| | | |
Collapse
|
46
|
Coelho CH, Tang WK, Burkhardt M, Galson JD, Muratova O, Salinas ND, Alves e Silva TL, Reiter K, MacDonald NJ, Nguyen V, Herrera R, Shimp R, Narum DL, Byrne-Steele M, Pan W, Hou X, Brown B, Eisenhower M, Han J, Jenkins BJ, Doritchamou JYA, Smelkinson MG, Vega-Rodríguez J, Trück J, Taylor JJ, Sagara I, Healy SA, Renn JP, Tolia NH, Duffy PE. A human monoclonal antibody blocks malaria transmission and defines a highly conserved neutralizing epitope on gametes. Nat Commun 2021; 12:1750. [PMID: 33741942 PMCID: PMC7979743 DOI: 10.1038/s41467-021-21955-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 02/17/2021] [Indexed: 01/31/2023] Open
Abstract
Malaria elimination requires tools that interrupt parasite transmission. Here, we characterize B cell receptor responses among Malian adults vaccinated against the first domain of the cysteine-rich 230 kDa gamete surface protein Pfs230, a key protein in sexual stage development of P. falciparum parasites. Among nine Pfs230 human monoclonal antibodies (mAbs) that we generated, one potently blocks transmission to mosquitoes in a complement-dependent manner and reacts to the gamete surface; the other eight show only low or no blocking activity. The structure of the transmission-blocking mAb in complex with vaccine antigen reveals a large discontinuous conformational epitope, specific to domain 1 of Pfs230 and comprising six structural elements in the protein. The epitope is conserved, suggesting the transmission-blocking mAb is broadly functional. This study provides a rational basis to improve malaria vaccines and develop therapeutic antibodies for malaria elimination.
Collapse
Affiliation(s)
- Camila H. Coelho
- grid.94365.3d0000 0001 2297 5165Pathogenesis and Immunity Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Wai Kwan Tang
- grid.94365.3d0000 0001 2297 5165Host-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
| | - Martin Burkhardt
- grid.94365.3d0000 0001 2297 5165Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Jacob D. Galson
- grid.7400.30000 0004 1937 0650Division of Immunology and Children’s Research Center, University Children’s Hospital Zurich, University of Zurich (UZH), Zurich, Switzerland ,Alchemab Therapeutics Ltd, 55-56 Russell Square, London, UK
| | - Olga Muratova
- grid.94365.3d0000 0001 2297 5165Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Nichole D. Salinas
- grid.94365.3d0000 0001 2297 5165Host-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
| | - Thiago Luiz Alves e Silva
- grid.94365.3d0000 0001 2297 5165Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD USA
| | - Karine Reiter
- grid.94365.3d0000 0001 2297 5165Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Nicholas J. MacDonald
- grid.94365.3d0000 0001 2297 5165Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Vu Nguyen
- grid.94365.3d0000 0001 2297 5165Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Raul Herrera
- grid.94365.3d0000 0001 2297 5165Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Richard Shimp
- grid.94365.3d0000 0001 2297 5165Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - David L. Narum
- grid.94365.3d0000 0001 2297 5165Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | | | - Wenjing Pan
- grid.429220.fiRepertoire Inc., Huntsville, AL USA
| | - Xiaohong Hou
- grid.429220.fiRepertoire Inc., Huntsville, AL USA
| | | | | | - Jian Han
- grid.429220.fiRepertoire Inc., Huntsville, AL USA
| | - Bethany J. Jenkins
- grid.94365.3d0000 0001 2297 5165Pathogenesis and Immunity Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Justin Y. A. Doritchamou
- grid.94365.3d0000 0001 2297 5165Pathogenesis and Immunity Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Margery G. Smelkinson
- grid.94365.3d0000 0001 2297 5165Biological Imaging Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Joel Vega-Rodríguez
- grid.94365.3d0000 0001 2297 5165Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD USA
| | - Johannes Trück
- grid.7400.30000 0004 1937 0650Division of Immunology and Children’s Research Center, University Children’s Hospital Zurich, University of Zurich (UZH), Zurich, Switzerland
| | - Justin J. Taylor
- grid.270240.30000 0001 2180 1622Fred Hutchinson Cancer Research Center, Seattle, WA USA
| | - Issaka Sagara
- Malaria Research and Training Center, University of Sciences, Techniques, and Technology, Bamako, Mali
| | - Sara A. Healy
- grid.94365.3d0000 0001 2297 5165Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Jonathan P. Renn
- grid.94365.3d0000 0001 2297 5165Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Niraj H. Tolia
- grid.94365.3d0000 0001 2297 5165Host-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
| | - Patrick E. Duffy
- grid.94365.3d0000 0001 2297 5165Pathogenesis and Immunity Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA ,grid.94365.3d0000 0001 2297 5165Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|
47
|
Chawla J, Oberstaller J, Adams JH. Targeting Gametocytes of the Malaria Parasite Plasmodium falciparum in a Functional Genomics Era: Next Steps. Pathogens 2021; 10:346. [PMID: 33809464 PMCID: PMC7999360 DOI: 10.3390/pathogens10030346] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 02/04/2023] Open
Abstract
Mosquito transmission of the deadly malaria parasite Plasmodium falciparum is mediated by mature sexual forms (gametocytes). Circulating in the vertebrate host, relatively few intraerythrocytic gametocytes are picked up during a bloodmeal to continue sexual development in the mosquito vector. Human-to-vector transmission thus represents an infection bottleneck in the parasite's life cycle for therapeutic interventions to prevent malaria. Even though recent progress has been made in the identification of genetic factors linked to gametocytogenesis, a plethora of genes essential for sexual-stage development are yet to be unraveled. In this review, we revisit P. falciparum transmission biology by discussing targetable features of gametocytes and provide a perspective on a forward-genetic approach for identification of novel transmission-blocking candidates in the future.
Collapse
Affiliation(s)
- Jyotsna Chawla
- Molecular Medicine, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, MDC 7, Tampa, FL 33612, USA;
| | - Jenna Oberstaller
- Center for Global Health and Infectious Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Suite 404, Tampa, FL 33612, USA;
| | - John H. Adams
- Center for Global Health and Infectious Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Suite 404, Tampa, FL 33612, USA;
| |
Collapse
|
48
|
Abstract
Introduction: An effective vaccine against malaria forms a global health priority. Both naturally acquired immunity and sterile protection induced by irradiated sporozoite immunization were described decades ago. Still no vaccine exists that sufficiently protects children in endemic areas. Identifying immunological correlates of vaccine efficacy can inform rational vaccine design and potentially accelerate clinical development.Areas covered: We discuss recent research on immunological correlates of malaria vaccine efficacy, including: insights from state-of-the-art omics platforms and systems vaccinology analyses; functional anti-parasitic assays; pre-immunization predictors of vaccine efficacy; and comparison of correlates of vaccine efficacy against controlled human malaria infections (CHMI) and against naturally acquired infections.Expert Opinion: Effective vaccination may be achievable without necessarily understanding immunological correlates, but the relatively disappointing efficacy of malaria vaccine candidates in target populations is concerning. Hypothesis-generating omics and systems vaccinology analyses, alongside assessment of pre-immunization correlates, have the potential to bring about paradigm-shifts in malaria vaccinology. Functional assays may represent in vivo effector mechanisms, but have scarcely been formally assessed as correlates. Crucially, evidence is still meager that correlates of vaccine efficacy against CHMI correspond with those against naturally acquired infections in target populations. Finally, the diversity of immunological assays and efficacy endpoints across malaria vaccine trials remains a major confounder.
Collapse
Affiliation(s)
| | - Matthew B B McCall
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, The Netherlands.,Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| |
Collapse
|
49
|
Lozano JM, Rodríguez Parra Z, Hernández-Martínez S, Yasnot-Acosta MF, Rojas AP, Marín-Waldo LS, Rincón JE. The Search of a Malaria Vaccine: The Time for Modified Immuno-Potentiating Probes. Vaccines (Basel) 2021; 9:vaccines9020115. [PMID: 33540947 PMCID: PMC7913233 DOI: 10.3390/vaccines9020115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/25/2022] Open
Abstract
Malaria is a deadly disease that takes the lives of more than 420,000 people a year and is responsible for more than 229 million clinical cases globally. In 2019, 95% of malaria morbidity occurred in African countries. The development of a highly protective vaccine is an urgent task that remains to be solved. Many vaccine candidates have been developed, from the use of the entire attenuated and irradiated pre-erythrocytic parasite forms (or recombinantly expressed antigens thereof) to synthetic candidates formulated in a variety of adjuvants and delivery systems, however these have unfortunately proven a limited efficacy. At present, some vaccine candidates are finishing safety and protective efficacy trials, such as the PfSPZ and the RTS,S/AS01 which are being introduced in Africa. We propose a strategy for introducing non-natural elements into target antigens representing key epitopes of Plasmodium spp. Accordingly, chemical strategies and knowledge of host immunity to Plasmodium spp. have served as the basis. Evidence is obtained after being tested in experimental rodent models for malaria infection and recognized for human sera from malaria-endemic regions. This encourages us to propose such an immune-potentiating strategy to be further considered in the search for new vaccine candidates.
Collapse
Affiliation(s)
- José Manuel Lozano
- Grupo de Investigación Mimetismo Molecular de los Agentes Infecciosos, Departamento de Farmacia, Universidad Nacional de Colombia—Sede Bogotá, 111321 Bogota, Colombia;
- Correspondence: ; Tel.: +57-3102-504-657
| | - Zully Rodríguez Parra
- Grupo de Investigación Mimetismo Molecular de los Agentes Infecciosos, Departamento de Farmacia, Universidad Nacional de Colombia—Sede Bogotá, 111321 Bogota, Colombia;
| | - Salvador Hernández-Martínez
- Dirección de Infección e Inmunidad, Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, 62508 Cuernavaca, Morelos, Mexico;
| | - Maria Fernanda Yasnot-Acosta
- Grupo de Investigaciones Microbiológicas y Biomédicas de Córdoba, Universidad de Córdoba, 230002 Monteria, Colombia;
| | - Angela Patricia Rojas
- Grupo de Investigación Biología Celular y Autoinmuniad, Departamento de Farmacia, Universidad Nacional de Colombia-Sede Bogotá, 111321 Bogota, Colombia;
| | | | - Juan Edilberto Rincón
- Departamento de Ingeniería y Mecatrónica, Universidad Nacional de Colombia-Sede Bogotá, 111321 Bogota, Colombia;
| |
Collapse
|
50
|
Duffy PE. Transmission-Blocking Vaccines: Harnessing Herd Immunity for Malaria Elimination. Expert Rev Vaccines 2021; 20:185-198. [PMID: 33478283 PMCID: PMC11127254 DOI: 10.1080/14760584.2021.1878028] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/14/2021] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Transmission-blocking vaccines (TBV) prevent community spread of malaria by targeting mosquito sexual stage parasites, a life-cycle bottleneck, and will be used in elimination programs. TBV rely on herd immunity to reduce mosquito infections and thereby new infections in both vaccine recipients and non-recipients, but do not provide protection once an individual receives an infectious mosquito bite which complicates clinical development. AREAS COVERED Here, we describe the concept and biology behind TBV, and we provide an update on clinical development of the leading vaccine candidate antigens. Search terms 'malaria vaccine,' 'sexual stages,' 'transmission blocking vaccine,' 'VIMT' and 'SSM-VIMT' were used for PubMed queries to identify relevant literature. EXPERT OPINION Candidates targeting P. falciparum zygote surface antigen Pfs25, and its P. vivax orthologue Pvs25, induced functional activity in humans that reduced mosquito infection in surrogate assays, but require increased durability to be useful in the field. Candidates targeting gamete surface antigens Pfs230 and Pfs48/45, respectively, are in or nearing clinical trials. Nanoparticle platforms and adjuvants are being explored to enhance immunogenicity. Efficacy trials require special considerations, such as cluster-randomized designs to measure herd immunity that reduces human and mosquito infection rates, while addressing human and mosquito movements as confounding factors.
Collapse
Affiliation(s)
- Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|