1
|
Xia M, Huang P, Vago FS, Jiang W, Jiang X, Tan M. A Nanoparticle Comprising the Receptor-Binding Domains of Norovirus and Plasmodium as a Combination Vaccine Candidate. Vaccines (Basel) 2025; 13:34. [PMID: 39852813 PMCID: PMC11769195 DOI: 10.3390/vaccines13010034] [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: 11/26/2024] [Revised: 12/23/2024] [Accepted: 12/28/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Noroviruses, which cause epidemic acute gastroenteritis, and Plasmodium parasites, which lead to malaria, are two infectious pathogens that pose threats to public health. The protruding (P) domain of norovirus VP1 and the αTSR domain of the circumsporozoite protein (CSP) of Plasmodium sporozoite are the glycan receptor-binding domains of the two pathogens for host cell attachment, making them excellent targets for vaccine development. Modified norovirus P domains self-assemble into a 24-meric octahedral P nanoparticle (P24 NP). METHODS We generated a unique P24-αTSR NP by inserting the αTSR domain into a surface loop of the P domain. The P-αTSR fusion proteins were produced in the Escherichia coli expression system and the fusion protein self-assembled into the P24-αTSR NP. RESULTS The formation of the P24-αTSR NP was demonstrated through gel filtration, electron microscopy, and dynamic light scattering. A 3D structural model of the P24-αTSR NP was constructed, using the known cryo-EM structure of the previously developed P24 NP and P24-VP8* NP as templates. Each P24-αTSR NP consists of a P24 NP core, with 24 surface-exposed αTSR domains that have retained their general conformations and binding function to heparan sulfate proteoglycans. The P24-αTSR NP is immunogenic, eliciting strong antibody responses in mice toward both the norovirus P domain and the αTSR domain of Plasmodium CSP. Notably, sera from mice immunized with the P24-αTSR NP bound strongly to Plasmodium sporozoites and blocked norovirus VLP attachment to their glycan receptors. CONCLUSION These data suggest that the P24-αTSR NP may serve as a combination vaccine against both norovirus and Plasmodium parasites.
Collapse
Affiliation(s)
- Ming Xia
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; (M.X.); (P.H.)
| | - Pengwei Huang
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; (M.X.); (P.H.)
| | - Frank S. Vago
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (F.S.V.); (W.J.)
| | - Wen Jiang
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (F.S.V.); (W.J.)
| | - Xi Jiang
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; (M.X.); (P.H.)
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Ming Tan
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; (M.X.); (P.H.)
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
2
|
Al-Osaimi HM, Kanan M, Marghlani L, Al-Rowaili B, Albalawi R, Saad A, Alasmari S, Althobaiti K, Alhulaili Z, Alanzi A, Alqarni R, Alsofiyani R, Shrwani R. A systematic review on malaria and dengue vaccines for the effective management of these mosquito borne diseases: Improving public health. Hum Vaccin Immunother 2024; 20:2337985. [PMID: 38602074 PMCID: PMC11017952 DOI: 10.1080/21645515.2024.2337985] [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: 02/17/2024] [Accepted: 03/28/2024] [Indexed: 04/12/2024] Open
Abstract
Insect vector-borne diseases (VBDs) pose significant global health challenges, particularly in tropical and subtropical regions. The WHO has launched the "Global Vector Control Response (GVCR) 2017-2030" to address these diseases, emphasizing a comprehensive approach to vector control. This systematic review investigates the potential of malaria and dengue vaccines in controlling mosquito-borne VBDs, aiming to alleviate disease burdens and enhance public health. Following PRISMA 2020 guidelines, the review incorporated 39 new studies out of 934 identified records. It encompasses various studies assessing malaria and dengue vaccines, emphasizing the significance of vaccination as a preventive measure. The findings indicate variations in vaccine efficacy, duration of protection, and safety considerations for each disease, influencing public health strategies. The review underscores the urgent need for vaccines to combat the increasing burden of VBDs like malaria and dengue, advocating for ongoing research and investment in vaccine development.
Collapse
Affiliation(s)
- Hind M. Al-Osaimi
- Department of Pharmacy Services Administration, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh, Kingdom of Saudi Arabia
| | - Mohammed Kanan
- Department of Clinical Pharmacy, King Fahad Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Lujain Marghlani
- Department of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia
| | - Badria Al-Rowaili
- Pharmaceutical Services Department, Northern Area Armed Forces Hospital, King Khalid Military, Hafr Al Batin, Kingdom of Saudi Arabia
| | - Reem Albalawi
- Department of Medicine, Tabuk University, Tabuk, Kingdom of Saudi Arabia
| | - Abrar Saad
- Pharmacy Department, Royal Commission Hospital, Yanbu, Kingdom of Saudi Arabia
| | - Saba Alasmari
- Department of Clinical Pharmacy, King Khalid University, Jeddah, Kingdom of Saudi Arabia
| | - Khaled Althobaiti
- Department of Medicine, Taif University, Ta’if, Kingdom of Saudi Arabia
| | - Zainab Alhulaili
- Department of Clinical Pharmacy, Dammam Medical Complex, Dammam, Kingdom of Saudi Arabia
| | - Abeer Alanzi
- Department of Medicine, King Abdulaziz Hospital, Makkah, Kingdom of Saudi Arabia
| | - Rawan Alqarni
- Department of Medicine and Surgery, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Razan Alsofiyani
- Department of Medicine, Taif University, Ta’if, Kingdom of Saudi Arabia
| | - Reem Shrwani
- Department of Clinical Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
3
|
Gbedande K, Ibitokou SA, Endrino MJD, Yap GS, Brown MG, Stephens R. Heightened innate immune state induced by viral vector leads to enhanced response to challenge and prolongs malaria vaccine protection. iScience 2024; 27:111468. [PMID: 39758993 PMCID: PMC11697717 DOI: 10.1016/j.isci.2024.111468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/01/2024] [Accepted: 11/21/2024] [Indexed: 01/07/2025] Open
Abstract
Cytomegalovirus is a promising vaccine vector; however, mechanisms promoting CD4 T cell responses to challenge, by CMV as a vector, are unknown. The ability of MCMV to prolong immunity generated by short-lived malaria vaccine was tested. MCMV provided non-specific protection to challenge with Plasmodium and increased interleukin-12 (IL-12) and CD8α+ dendritic cell (DC) numbers through prolonged MCMV-dependent interferon gamma (IFN-γ) production. This late innate response to MCMV increased IL-12 upon challenge and increased the polyclonal CD4 effector T cell response to Plasmodium, protecting in an IL-12-dependent manner. Although Plasmodium-vaccine-induced protection decayed by d200, MCMV restored protection through IFN-γ. Mechanistically, protection depended on MCMV-induced-IFN-γ increasing CD8α+ DCs and IL-12p40. MCMV expressing a Plasmodium epitope increased parasite-specific CD4 effector and effector memory T cells persisting after malaria vaccination, both phenotypes reported to protect. Overall, enhanced innate cell status, a mechanism of heterologous protection by MCMV, led to a stronger T cell response to challenge.
Collapse
Affiliation(s)
- Komi Gbedande
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555-0435, USA
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Cancer Center, 205 S. Orange Avenue, Newark, NJ 07103, USA
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Cancer Center, 205 S. Orange Avenue, Newark, NJ 07103, USA
| | - Samad A. Ibitokou
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555-0435, USA
| | - Mark Joseph D. Endrino
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Cancer Center, 205 S. Orange Avenue, Newark, NJ 07103, USA
| | - George S. Yap
- Center for Immunity and Inflammation, and Department of Medicine, Rutgers New Jersey Medical School, Cancer Center, 205 S. Orange Avenue, Newark, NJ 07103, USA
| | - Michael G. Brown
- Department of Medicine, Division of Nephrology, and the Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
| | - Robin Stephens
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555-0435, USA
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Cancer Center, 205 S. Orange Avenue, Newark, NJ 07103, USA
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Cancer Center, 205 S. Orange Avenue, Newark, NJ 07103, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
4
|
Plieskatt J, Ofori EA, Naghizadeh M, Miura K, Flores-Garcia Y, Borbye-Lorenzen N, Tiono AB, Skogstrand K, Sagara I, Zavala F, Theisen M. ProC6C, a novel multi-stage malaria vaccine, elicits functional antibodies against the minor and central repeats of the Circumsporozoite Protein in human adults. Front Immunol 2024; 15:1481829. [PMID: 39555079 PMCID: PMC11563800 DOI: 10.3389/fimmu.2024.1481829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/11/2024] [Indexed: 11/19/2024] Open
Abstract
Introduction ProC6C is a multi-stage malaria vaccine which includes Plasmodium falciparum Circumsporozoite Protein (PfCSP), Pfs48/45 and Pfs230 sequences, designed to elicit functional antibodies that prevent sporozoite invasion of human hepatocytes (PfCSP) and parasite development in mosquitoes (Pfs48/45 and Pfs230). ProC6C formulated on Alhydrogel was evaluated in combination with Matrix-M in a Phase 1 trial in Burkina Faso. The PfCSP antibody responses were assessed for magnitude, specificity, avidity and functionality. These results compliment the prior reported safety and tolerability of ProC6C as well as the transmission reducing activity of ProC6C. Methods The PfCSP response of ProC6C in Burkinabes in the Phase 1 trial (PACTR202201848463189) was profiled through the three vaccine administrations of 100 µg protein on Alhydrogel® alone (AlOH) or combined with 50 µg Matrix-M™ adjuvant (AlOH/Matrix-M). Serology was completed against full-length PfCSP and major/minor repeat peptides using antibody equivalence to PfCSP monoclonal antibodies (mAb 311, mAb 317 and mAb L9). Comparison of the ProC6C responses were made to those that received RTS,S/AS01 in a study conducted in Thailand. Bio-Layer Interferometry was further used to determine antibody avidity. The human IgG was subsequently purified, pooled, and evaluated in a mouse sporozoite challenge model to determine functionality. Results A single administration of ProC6C-AlOH/Matrix-M seroconverted 19 of 20 volunteers against PfCSP and significantly enhanced antibody titers to major and minor repeats (and present through D180). At D70, ProC6C-AlOH/Matrix-M PfCSP antibodies were found to be similar to responder pools generated from Thai adults receiving RTS,S/AS01. Additionally, ProC6C antibodies were found to be competitive to established PfCSP antibodies such as mAb 317 and mAb L9. The purified and pooled IgG from human volunteers, used in a passive transfer mouse sporozoite challenge model, showed a median of 50% inhibition (P=0.0058). ProC6C PfCSP antibodies were functional in this in vivo assessment and consistent with inhibition seen by other Circumsporozoite vaccines in this model. Discussion This analysis supports continued investigation of the antibody responses elicited by the ProC6C multi-stage malaria vaccine. This Phase 1 clinical trial demonstrated the short PfCSP sequence included in ProC6C can induce significant PfCSP antibodies in humans, which importantly were determined to be functional.
Collapse
Affiliation(s)
- Jordan Plieskatt
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
| | - Ebenezer Addo Ofori
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
- Centre for Translational Medicine and Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Mohammad Naghizadeh
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
- Centre for Translational Medicine and Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Nis Borbye-Lorenzen
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
| | - Alfred B. Tiono
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Kristin Skogstrand
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
| | - Issaka Sagara
- Malaria Research and Training Center, Mali- National Institute of Allergy and Infectious Diseases International Center for Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
- Centre for Translational Medicine and Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
5
|
Chutiyami M. Recent Trends in Malaria Vaccine Research Globally: A Bibliometric Analysis From 2005 to 2022. J Parasitol Res 2024; 2024:8201097. [PMID: 39483206 PMCID: PMC11527547 DOI: 10.1155/2024/8201097] [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: 03/01/2024] [Revised: 09/09/2024] [Accepted: 10/03/2024] [Indexed: 11/03/2024] Open
Abstract
Aim: Malaria vaccine is one of the critical areas in tropical health research, considering the success recorded in other vaccine-preventable diseases. This study is aimed at reviewing recent trends in global malaria vaccine research from 2005 to 2022. Method: A validated search strategy was undertaken to identify scientific literature on the malaria vaccine in the Scopus database. Bibliometric indicators identified include a pattern of publication growth and citations over the study period; top authors, countries, funding organizations, and journals; keywords, including different malarial parasite species, and the overall research themes. Result: A total of 6457 documents were found from 2005 to 2022, published in 160 journals/sources in 189 countries/territories. Malaria Journal published the highest number of research outputs (478, 7.4%) within the study period, and the highest number of documents (468, 7.3%) were published in 2021. There were 214,323 total citations, with 33.2 average citations per document and 167 documents' h-index. The United States, United Kingdom, and Australia combined produced more than 60% of the publication output, with most collaboration with African countries such as Kenya. Plasmodium falciparum is the most occurring parasite species keyword (754, 11.7%), with a growing interest in Plasmodium knowlesi (30, 0.5%). Merozoite surface protein, characterization, trials, infant/children, traveler, and research/review were the six themes that emerged from the studies. Conclusion: The last one and half decades have seen a significant increase in malaria vaccine research and citations, mainly targeting vaccine development, safety, and efficacy in Africa. This necessitates more international efforts to improve the vaccines' effectiveness considering different Plasmodium species.
Collapse
Affiliation(s)
- Muhammad Chutiyami
- School of Nursing and Midwifery, Faculty of Health, University of Technology Sydney, Sydney, Australia
| |
Collapse
|
6
|
Mishra A, Paul P, Srivastava M, Mishra S. A Plasmodium late liver stage arresting GAP provides superior protection in mice. NPJ Vaccines 2024; 9:193. [PMID: 39424860 PMCID: PMC11489731 DOI: 10.1038/s41541-024-00975-0] [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: 05/01/2024] [Accepted: 09/19/2024] [Indexed: 10/21/2024] Open
Abstract
Liver-stage genetically attenuated malaria parasites (GAPs) are powerful immunogens that provide protection against sporozoite challenge. We previously generated two late liver-stage-arresting GAPs by deleting the stearoyl-CoA desaturase (Scd) or sporozoite conserved orthologous transcript 1 (Scot1) genes in Plasmodium berghei. Immunization with Scd or Scot1 GAP conferred complete protection against a sporozoite challenge. In a safety study, we observed rare breakthrough blood-stage infections in mice inoculated with high doses of sporozoites, indicating that both GAPs were incompletely attenuated. In this study, we generated a Scd/Scot1 GAP by dual gene deletion. This resulted in complete attenuation of the parasites in the liver and did not transition to blood-stage infection despite a high-dose sporozoite challenge. The Scd/Scot1 KO and WT GFP parasites were indistinguishable during blood, mosquito and early liver stage development. Moreover, Scd/Scot1 KO liver-stage schizonts exhibited an abnormal apicoplast biogenesis and nuclear division phenotype, failed to form hepatic merozoites, and exhibited late liver-stage arrest. Compared with early-arresting Speld KO immunization, late-stage liver-arresting Scd/Scot1 KO induces greater and broader CD8+ T-cell responses and elicits stage-transcending immunity that provides superior protection in C57BL/6 mice. These data prove that multiple gene deletions lead to complete attenuation of the parasite and support the development of late liver stage-arresting P. falciparum GAP.
Collapse
Affiliation(s)
- Akancha Mishra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Plabita Paul
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Mrigank Srivastava
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Satish Mishra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
7
|
Ba A, Thiam LG, Pouye MN, Guo Y, Patel SD, Sene SD, Diallo F, Li R, Cisse A, Guerra N, Laqqa S, Mangou K, Moore AJ, Sadio BD, Ndiaye JLA, Mbengue A, Sheng Z, Shapiro L, Bei AK. Genetic diversity in the Plasmodium falciparum next-generation blood stage vaccine candidate antigen PfCyRPA in Senegal. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.13.24305808. [PMID: 39484257 PMCID: PMC11527082 DOI: 10.1101/2024.10.13.24305808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The Plasmodium falciparum cysteine-rich protective antigen (PfCyRPA) is a promising target as a next-generation blood-stage malaria vaccine and together with PCRCR complex members, the reticulocyte binding-like homologous protein 5 (PfRh5) and the Rh5-interacting protein (PfRipr), are currently being evaluated in clinical trials. PfCyRPA is essential for merozoite invasion and appears to be highly conserved within the P. falciparum parasite populations. Here, we used a targeted deep amplicon next-generation sequencing approach to assess the breadth of PfCyRPA genetic diversity in 95 P. falciparum clinical isolates from Kédougou, an area with a high seasonal malaria transmission in Senegal. Our data show the dominant prevalence of PfCyRPA wild type reference allele, while we also identify a total of 15 single nucleotide polymorphisms (SNPs). Of these, only five have previously been reported, while the majority of the SNPs were present as singletons within our sampled population. Moreover, the variant read frequency of the identified SNPs varied from 2.6 to 100%, while the majority of the SNPs were present at frequencies greater than 25% in polygenomic samples. We also applied a structure-based modelling approach to thread these SNPs onto PfCyRPA crystal structures and showed that these polymorphisms have different predicted functional impacts on the interactions with binding partner PfRH5 or neutralizing antibodies. Our prediction revealed that the majority of these SNPs have minor effects on PfCyRPA antibodies, while others alter its structure, stability, or interaction with PfRH5. Altogether, our present findings reveal conserved PfCyRPA epitopes which will inform downstream investigations on next-generation structure-guided malaria vaccine design.
Collapse
Affiliation(s)
- Aboubacar Ba
- G4 Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Laty Gaye Thiam
- G4 Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Mariama Nicole Pouye
- G4 Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Yicheng Guo
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Saurabh D Patel
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Seynabou Diouf Sene
- G4 Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Fatoumata Diallo
- G4 Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Rebecca Li
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Awa Cisse
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Noemi Guerra
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Safia Laqqa
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Khadidjatou Mangou
- G4 Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Adam J Moore
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Bacary Djilocalisse Sadio
- G4 Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | | | - Alassane Mbengue
- G4 Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
| | - Zizhang Sheng
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Lawrence Shapiro
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Amy K Bei
- G4 Malaria Experimental Genetic Approaches & Vaccines, Pôle Immunophysiopathologie et Maladies Infectieuses, Institut Pasteur de Dakar, Dakar, Senegal
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| |
Collapse
|
8
|
Chutiyami M, Saravanakumar P, Bello UM, Salihu D, Adeleye K, Kolo MA, Dawa KK, Hamina D, Bhandari P, Sulaiman SK, Sim J. Malaria vaccine efficacy, safety, and community perception in Africa: a scoping review of recent empirical studies. Infection 2024; 52:2007-2028. [PMID: 38441731 PMCID: PMC11499420 DOI: 10.1007/s15010-024-02196-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/22/2024] [Indexed: 10/24/2024]
Abstract
AIM The review summarizes the recent empirical evidence on the efficacy, safety, and community perception of malaria vaccines in Africa. METHODS Academic Search Complete, African Journals Online, CINAHL, Medline, PsychInfo, and two gray literature sources were searched in January 2023, and updated in June 2023. Relevant studies published from 2012 were included. Studies were screened, appraised, and synthesized in line with the review aim. Statistical results are presented as 95% Confidence Intervals and proportions/percentages. RESULTS Sixty-six (N = 66) studies met the inclusion criteria. Of the vaccines identified, overall efficacy at 12 months was highest for the R21 vaccine (N = 3) at 77.0%, compared to the RTS,S vaccine (N = 15) at 55%. The efficacy of other vaccines was BK-SE36 (11.0-50.0%, N = 1), ChAd63/MVA ME-TRAP (- 4.7-19.4%, N = 2), FMP2.1/AS02A (7.6-9.9%, N = 1), GMZ2 (0.6-60.0%, N = 5), PfPZ (20.0-100.0%, N = 5), and PfSPZ-CVac (24.8-33.6%, N = 1). Injection site pain and fever were the most common adverse events (N = 26), while febrile convulsion (N = 8) was the most reported, vaccine-related Serious Adverse Event. Mixed perceptions of malaria vaccines were found in African communities (N = 17); awareness was generally low, ranging from 11% in Tanzania to 60% in Nigeria (N = 9), compared to willingness to accept the vaccines, which varied from 32.3% in Ethiopia to 96% in Sierra Leone (N = 15). Other issues include availability, logistics, and misconceptions. CONCLUSION Malaria vaccines protect against malaria infection in varying degrees, with severe side effects rarely occurring. Further research is required to improve vaccine efficacy and community involvement is needed to ensure successful widespread use in African communities.
Collapse
Affiliation(s)
- Muhammad Chutiyami
- School of Nursing and Midwifery, University of Technology Sydney, Sydney, Australia.
| | - Priya Saravanakumar
- School of Nursing and Midwifery, University of Technology Sydney, Sydney, Australia
| | - Umar Muhammad Bello
- Department of Physiotherapy and Paramedicine, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Dauda Salihu
- College of Nursing, Jouf University, Sakaka, Saudi Arabia
| | - Khadijat Adeleye
- College of Nursing, University of Massachusetts, Amherst, MA, 01003, USA
| | | | - Kabiru Kasamu Dawa
- School of Nursing, Midwifery and Health Education, University of Bedfordshire, Luton, UK
| | - Dathini Hamina
- Department of Nursing Science, University of Maiduguri, Maiduguri, Nigeria
| | - Pratibha Bhandari
- School of Nursing and Midwifery, University of Technology Sydney, Sydney, Australia
| | | | - Jenny Sim
- WHO Collaborating Centre for Nursing, Midwifery and Health Development, University of Technology Sydney, Sydney, Australia
- School of Nursing, Midwifery and Paramedicine, Australian Catholic University, Sydney, Australia
| |
Collapse
|
9
|
Boyle MJ, Engwerda CR, Jagannathan P. The impact of Plasmodium-driven immunoregulatory networks on immunity to malaria. Nat Rev Immunol 2024; 24:637-653. [PMID: 38862638 PMCID: PMC11688169 DOI: 10.1038/s41577-024-01041-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 06/13/2024]
Abstract
Malaria, caused by infection with Plasmodium parasites, drives multiple regulatory responses across the immune landscape. These regulatory responses help to protect against inflammatory disease but may in some situations hamper the acquisition of adaptive immune responses that clear parasites. In addition, the regulatory responses that occur during Plasmodium infection may negatively affect malaria vaccine efficacy in the most at-risk populations. Here, we discuss the specific cellular mechanisms of immunoregulatory networks that develop during malaria, with a focus on knowledge gained from human studies and studies that involve the main malaria parasite to affect humans, Plasmodium falciparum. Leveraging this knowledge may lead to the development of new therapeutic approaches to increase protective immunity to malaria during infection or after vaccination.
Collapse
Affiliation(s)
- Michelle J Boyle
- Life Sciences Division, Burnet Institute, Melbourne, Victoria, Australia.
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
| | | | - Prasanna Jagannathan
- Department of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
10
|
Ranjha R, Bai P, Singh K, Mohan M, Bharti PK, Anvikar AR. Rethinking malaria vaccines: perspectives on currently approved malaria vaccines in India's path to elimination. BMJ Glob Health 2024; 9:e016019. [PMID: 39182925 PMCID: PMC11404236 DOI: 10.1136/bmjgh-2024-016019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 08/01/2024] [Indexed: 08/27/2024] Open
Affiliation(s)
- Ritesh Ranjha
- ICMR- National Institute of Malaria Research, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Priyanka Bai
- ICMR- National Institute of Malaria Research, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Kuldeep Singh
- ICMR- National Institute of Malaria Research, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Mradul Mohan
- ICMR- National Institute of Malaria Research, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Praveen K Bharti
- ICMR- National Institute of Malaria Research, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Anup R Anvikar
- ICMR- National Institute of Malaria Research, New Delhi, India
| |
Collapse
|
11
|
Zhou Y, Hatzakis K, MacMillen Z, Laohajaratsang M, Grieser AM, Itsara LS, Do J, Davie JW, Ghosh AK, Avril M. Full maturation of in vitro Plasmodium falciparum oocysts using the AlgiMatrix 3D culture system. Malar J 2024; 23:251. [PMID: 39164764 PMCID: PMC11334386 DOI: 10.1186/s12936-024-05079-7] [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: 05/29/2024] [Accepted: 08/13/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Plasmodium falciparum oocysts undergo growth and maturation in a unique setting within the mosquito midgut, firmly situated between the epithelium and the basal lamina. This location exposes them to specific nutrient exchange and metabolic processes while in direct contact with the mosquito haemolymph. The limited availability of in vitro culture systems for growth of the various P. falciparum mosquito stages hampers study of their biology and impedes progress in combatting malaria. METHODS An artificial in vitro environment was established to mimic this distinctive setting, transitioning from a 2D culture system to a 3D model capable of generating fully mature oocysts that give rise to in vitro sporozoites. RESULTS A two-dimensional (2D) chamber slide was employed along with an extracellular matrix composed of type IV collagen, entactin, and gamma laminin. This matrix facilitated development of the optimal medium composition for cultivating mature P. falciparum oocysts in vitro. However, the limitations of this 2D culture system in replicating the in vivo oocyst environment prompted a refinement of the approach by optimizing a three-dimensional (3D) alginate matrix culture system. This new system offered improved attachment, structural support, and nutrient exchange for the developing oocysts, leading to their maturation and the generation of sporozoites. CONCLUSIONS This technique enables the in vitro growth of P. falciparum oocysts and sporozoites.
Collapse
Affiliation(s)
- Yaxian Zhou
- MalarVx, Inc, 1551 Eastlake Ave N, Suite 100, Seattle, WA, 98102, USA
- University of Wisconsin-Madison, Madison, WI, USA
| | - Kiara Hatzakis
- MalarVx, Inc, 1551 Eastlake Ave N, Suite 100, Seattle, WA, 98102, USA
- HDT Bio, Seattle, WA, USA
| | - Zachary MacMillen
- MalarVx, Inc, 1551 Eastlake Ave N, Suite 100, Seattle, WA, 98102, USA
| | - Mint Laohajaratsang
- MalarVx, Inc, 1551 Eastlake Ave N, Suite 100, Seattle, WA, 98102, USA
- University of Washington, Seattle, WA, USA
| | - Alexis M Grieser
- MalarVx, Inc, 1551 Eastlake Ave N, Suite 100, Seattle, WA, 98102, USA
- College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Leslie S Itsara
- MalarVx, Inc, 1551 Eastlake Ave N, Suite 100, Seattle, WA, 98102, USA
- Adaptive Biotechnologies Corp, Seattle, USA
| | - Julie Do
- MalarVx, Inc, 1551 Eastlake Ave N, Suite 100, Seattle, WA, 98102, USA
- Institute for Systems Biology, Seattle, WA, USA
| | - James W Davie
- MalarVx, Inc, 1551 Eastlake Ave N, Suite 100, Seattle, WA, 98102, USA
| | - Anil K Ghosh
- MalarVx, Inc, 1551 Eastlake Ave N, Suite 100, Seattle, WA, 98102, USA
| | - Marion Avril
- MalarVx, Inc, 1551 Eastlake Ave N, Suite 100, Seattle, WA, 98102, USA.
| |
Collapse
|
12
|
Hanboonkunupakarn B, Mukaka M, Jittamala P, Poovorawan K, Pongsuwan P, Stockdale L, Provstgaard-Morys S, Chotivanich K, Tarning J, Hoglund RM, Chimjinda N, Ewer K, Ramos-Lopez F, Day NPJ, Dondorp AM, Hill AV, White NJ, von Seidlein L, Pukrittayakamee S. A randomised trial of malaria vaccine R21/Matrix-M™ with and without antimalarial drugs in Thai adults. NPJ Vaccines 2024; 9:124. [PMID: 38971837 PMCID: PMC11227592 DOI: 10.1038/s41541-024-00920-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/27/2024] [Indexed: 07/08/2024] Open
Abstract
In preparation for mass vaccinations with R21/Matrix-M™ combined with mass administrations of dihydroartemisinin, piperaquine, and a single low dose primaquine we assessed the tolerability, safety, and potential interactions of this combination affecting immunogenicity or pharmacokinetics. 120 healthy Thai volunteers were randomised to receive either antimalarials combined with vaccinations (n = 50), vaccinations alone (n = 50), or antimalarials only (n = 20). Three rounds of vaccines and antimalarials were administered one month apart. The vaccine was well tolerated alone and in combination with the antimalarials. None of the participants failed completion of the 3-dose vaccine course. There was no significant difference in the vaccine immunogenicity or in the pharmacokinetics of piperaquine given individually or in combination. This study supports proceeding to a large trial of mass vaccinations with R21/Matrix-M™ combined with mass antimalarial administration in Bangladesh.
Collapse
Affiliation(s)
- Borimas Hanboonkunupakarn
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Mavuto Mukaka
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Podjanee Jittamala
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kittiyod Poovorawan
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Pongphaya Pongsuwan
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Lisa Stockdale
- The Jenner Institute Laboratories, University of Oxford, Oxford, UK
| | | | - Kesinee Chotivanich
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Joel Tarning
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Richard M Hoglund
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Natenapa Chimjinda
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Katie Ewer
- The Jenner Institute Laboratories, University of Oxford, Oxford, UK
- GSK, GSK Vaccines Institute for Global Health, Siena, Italy
| | - Fernando Ramos-Lopez
- Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Nicholas P J Day
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Arjen M Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Adrian V Hill
- The Jenner Institute Laboratories, University of Oxford, Oxford, UK
| | - Nicholas J White
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Lorenz von Seidlein
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK.
| | - Sasithon Pukrittayakamee
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- The Royal Society of Thailand, Dusit, Bangkok, Thailand
| |
Collapse
|
13
|
El-Moamly AA. How can we get malaria control back on track? BMJ 2024; 385:q1408. [PMID: 38925798 DOI: 10.1136/bmj.q1408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Affiliation(s)
- Amal A El-Moamly
- Department of Medical Parasitology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
14
|
Gouleu CS, Daouda MA, Oye Bingono SO, McCall MBB, Alabi AS, Adegnika AA, Schaumburg F, Grebe T. Temporal trends of skin and soft tissue infections caused by methicillin-resistant Staphylococcus aureus in Gabon. Antimicrob Resist Infect Control 2024; 13:68. [PMID: 38918863 PMCID: PMC11201302 DOI: 10.1186/s13756-024-01426-0] [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: 02/13/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Methicillin-resistant Staphylococcus aureus (MRSA) is one of the leading causes of mortality due to bacterial antimicrobial resistance. While S. aureus is common in skin and soft tissue infections (SSTI) in Africa, data on MRSA rates are scarce and reports vary widely across the continent (5%-80%). In this study, we describe the proportion of MRSA causing SSTI in Lambaréné, Gabon, over an 11-year period. METHODS We retrospectively analyzed data from 953 bacterial specimens collected from inpatients and outpatients with SSTI at the Albert Schweitzer Hospital, Lambaréné, Gabon, between 2009 and 2019. We determined temporal changes in the prevalence of MRSA and identified risk factors for SSTI with MRSA. RESULTS 68% of all specimens with bacterial growth yielded S. aureus (n = 499/731), of which 7% (36/497) with antimicrobial susceptibility testing were identified as MRSA. Age above 18 years, admission to the surgical ward, and deep-seated infections were significantly associated with MRSA as the causative agent. After an initial decline from 7% in 2009, there was a marked increase in the proportion of MRSA among all S. aureus from SSTI from 3 to 20% between 2012 and 2019. The resistance rate to erythromycin was significantly higher in MRSA than in methicillin-susceptible S. aureus (73% vs. 10%), and clindamycin resistance was detected exclusively in MRSA isolates (8%). CONCLUSION The increasing proportion of MRSA causing SSTI over the 11-year period contrasts with many European countries where MRSA is on decline. Continuous surveillance of MRSA lineages in the hospital and community along with antibiotic stewardship programs could address the increasing trend of MRSA.
Collapse
Affiliation(s)
| | | | | | - Matthew Benjamin Bransby McCall
- Radboud Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Ayola Akim Adegnika
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institute of Tropical Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Frieder Schaumburg
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Tobias Grebe
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany.
| |
Collapse
|
15
|
Wistuba-Hamprecht J, Reuter B, Fendel R, Hoffman SL, Campo JJ, Felgner PL, Kremsner PG, Mordmüller B, Pfeifer N. Machine learning prediction of malaria vaccine efficacy based on antibody profiles. PLoS Comput Biol 2024; 20:e1012131. [PMID: 38848436 PMCID: PMC11189177 DOI: 10.1371/journal.pcbi.1012131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 06/20/2024] [Accepted: 05/02/2024] [Indexed: 06/09/2024] Open
Abstract
Immunization through repeated direct venous inoculation of Plasmodium falciparum (Pf) sporozoites (PfSPZ) under chloroquine chemoprophylaxis, using the PfSPZ Chemoprophylaxis Vaccine (PfSPZ-CVac), induces high-level protection against controlled human malaria infection (CHMI). Humoral and cellular immunity contribute to vaccine efficacy but only limited information about the implicated Pf-specific antigens is available. Here, we examined Pf-specific antibody profiles, measured by protein arrays representing the full Pf proteome, of 40 placebo- and PfSPZ-immunized malaria-naïve volunteers from an earlier published PfSPZ-CVac dose-escalation trial. For this purpose, we both utilized and adapted supervised machine learning methods to identify predictive antibody profiles at two different time points: after immunization and before CHMI. We developed an adapted multitask support vector machine (SVM) approach and compared it to standard methods, i.e. single-task SVM, regularized logistic regression and random forests. Our results show, that the multitask SVM approach improved the classification performance to discriminate the protection status based on the underlying antibody-profiles while combining time- and dose-dependent data in the prediction model. Additionally, we developed the new fEature diStance exPlainabilitY (ESPY) method to quantify the impact of single antigens on the non-linear multitask SVM model and make it more interpretable. In conclusion, our multitask SVM model outperforms the studied standard approaches in regard of classification performance. Moreover, with our new explanation method ESPY, we were able to interpret the impact of Pf-specific antigen antibody responses that predict sterile protective immunity against CHMI after immunization. The identified Pf-specific antigens may contribute to a better understanding of immunity against human malaria and may foster vaccine development.
Collapse
Affiliation(s)
- Jacqueline Wistuba-Hamprecht
- Department of Computer Science, University of Tübingen, Tübingen, Germany
- Institute for Biomedical Informatics, University of Tübingen, Tübingen, Germany
| | - Bernhard Reuter
- Department of Computer Science, University of Tübingen, Tübingen, Germany
- Institute for Biomedical Informatics, University of Tübingen, Tübingen, Germany
| | - Rolf Fendel
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, partner site Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | | | - Joseph J. Campo
- Antigen Discovery Inc., Irvine, California, United States of America
| | - Philip L. Felgner
- Department of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Peter G. Kremsner
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, partner site Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Benjamin Mordmüller
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nico Pfeifer
- Department of Computer Science, University of Tübingen, Tübingen, Germany
- Institute for Biomedical Informatics, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, partner site Tübingen, Tübingen, Germany
| |
Collapse
|
16
|
Kusi KA, Amoah LE, Acquah FK, Ennuson NA, Frempong AF, Ofori EA, Akyea-Mensah K, Kyei-Baafour E, Osei F, Frimpong A, Singh SK, Theisen M, Remarque EJ, Faber BW, Belmonte M, Ganeshan H, Huang J, Villasante E, Sedegah M. Plasmodium falciparum AMA1 and CSP antigen diversity in parasite isolates from southern Ghana. Front Cell Infect Microbiol 2024; 14:1375249. [PMID: 38808064 PMCID: PMC11132687 DOI: 10.3389/fcimb.2024.1375249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/19/2024] [Indexed: 05/30/2024] Open
Abstract
Introduction Diversity in malarial antigens is an immune evasion mechanism that gives malaria parasites an edge over the host. Immune responses against one variant of a polymorphic antigen are usually not fully effective against other variants due to altered epitopes. This study aimed to evaluate diversity in the Plasmodium falciparum antigens apical membrane antigen 1 (PfAMA1) and circumsporozoite protein (PfCSP) from circulating parasites in a malaria-endemic community in southern Ghana and to determine the effects of polymorphisms on antibody response specificity. Methods The study involved 300 subjects, whose P. falciparum infection status was determined by microscopy and PCR. Diversity within the two antigens was evaluated by msp2 gene typing and molecular gene sequencing, while the host plasma levels of antibodies against PfAMA1, PfCSP, and two synthetic 24mer peptides from the conserved central repeat region of PfCSP, were measured by ELISA. Results Of the 300 subjects, 171 (57%) had P. falciparum infection, with 165 of the 171 (96.5%) being positive for either or both of the msp2 allelic families. Gene sequencing of DNA from 55 clonally infected samples identified a total of 56 non-synonymous single nucleotide polymorphisms (SNPs) for the Pfama1 gene and these resulted in 44 polymorphic positions, including two novel positions (363 and 365). Sequencing of the Pfcsp gene from 69 clonal DNA samples identified 50 non-synonymous SNPs that resulted in 42 polymorphic positions, with half (21) of these polymorphic positions being novel. Of the measured antibodies, only anti-PfCSP antibodies varied considerably between PCR parasite-positive and parasite-negative persons. Discussion These data confirm the presence of a considerable amount of unique, previously unreported amino acid changes, especially within PfCSP. Drivers for this diversity in the Pfcsp gene do not immediately seem apparent, as immune pressure will be expected to drive a similar level of diversity in the Pfama1 gene.
Collapse
Affiliation(s)
- Kwadwo A. Kusi
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Linda E. Amoah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Festus Kojo Acquah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Nana Aba Ennuson
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Abena F. Frempong
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Ebenezer A. Ofori
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Kwadwo Akyea-Mensah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Eric Kyei-Baafour
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Frank Osei
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Augustina Frimpong
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Susheel K. Singh
- Center for Medical Parasitology at the Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Congenital Diseases, Statens Serum Institut, Copenhagen, Denmark
| | - Michael Theisen
- Center for Medical Parasitology at the Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Congenital Diseases, Statens Serum Institut, Copenhagen, Denmark
| | - Edmond J. Remarque
- Department of Parasitology, Biomedical Primate Research Center, Rijswijk, Netherlands
| | - Bart W. Faber
- Department of Parasitology, Biomedical Primate Research Center, Rijswijk, Netherlands
| | - Maria Belmonte
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Malaria Department, Naval Medical Research Command, Silver Spring, MD, United States
| | - Harini Ganeshan
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Malaria Department, Naval Medical Research Command, Silver Spring, MD, United States
| | - Jun Huang
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Malaria Department, Naval Medical Research Command, Silver Spring, MD, United States
| | - Eileen Villasante
- Malaria Department, Naval Medical Research Command, Silver Spring, MD, United States
| | - Martha Sedegah
- Malaria Department, Naval Medical Research Command, Silver Spring, MD, United States
| |
Collapse
|
17
|
Osoro CB, Ochodo E, Kwambai TK, Otieno JA, Were L, Sagam CK, Owino EJ, Kariuki S, Ter Kuile FO, Hill J. Policy uptake and implementation of the RTS,S/AS01 malaria vaccine in sub-Saharan African countries: status 2 years following the WHO recommendation. BMJ Glob Health 2024; 9:e014719. [PMID: 38688566 PMCID: PMC11085798 DOI: 10.1136/bmjgh-2023-014719] [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: 12/01/2023] [Accepted: 03/31/2024] [Indexed: 05/02/2024] Open
Abstract
In October 2021, the WHO recommended the world's first malaria vaccine-RTS,S/AS01-to prevent malaria in children living in areas with moderate-to-high transmission in sub-Saharan Africa (SSA). A second malaria vaccine, R21/Matrix-M, was recommended for use in October 2023 and added to the WHO list of prequalified vaccines in December 2023. This study analysis assessed the country status of implementation and delivery strategies for RTS,S/AS01 by searching websites for national malaria policies, guidelines and related documents. Direct contact with individuals working in malaria programmes was made to obtain documents not publicly available. 10 countries had documents with information relating to malaria vaccine implementation, 7 referencing RTS,S/AS01 and 3 (Burkina Faso, Kenya and Nigeria) referencing RTS,S/AS01 and R21/Matrix-M. Five other countries reported plans for malaria vaccine roll-out without specifying which vaccine. Ghana, Kenya and Malawi, which piloted RTS,S/AS01, have now integrated the vaccine into routine immunisation services. Cameroon and Burkina Faso are the first countries outside the pilot countries to incorporate the vaccine into national immunisation services. Uganda plans a phased RTS,S/AS01 introduction, while Guinea plans to first pilot RTS,S/AS01 in five districts. The RTS,S/AS01 schedule varied by country, with the first dose administered at 5 or 6 months in all countries but the fourth dose at either 18, 22 or 24 months. SSA countries have shown widespread interest in rolling out the malaria vaccine, the Global Alliance for Vaccines and Immunization having approved financial support for 20 of 30 countries which applied as of March 2024. Limited availability of RTS,S/AS01 means that some approved countries will not receive the required doses. Vaccine availability and equity must be addressed even as R21/Matrix-M becomes available.
Collapse
Affiliation(s)
- Caroline Bonareri Osoro
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
- Department of Global Health, Stellenbosch University, Stellenbosch, South Africa
| | - Eleanor Ochodo
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
- Department of Global Health, Stellenbosch University, Stellenbosch, South Africa
| | | | - Jenifer Akoth Otieno
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Lisa Were
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Caleb Kimutai Sagam
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Eddy Johnson Owino
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Simon Kariuki
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Feiko O Ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Jenny Hill
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
18
|
Dobbs KR, Atieli HE, Valim C, Beeson JG. Previous Malaria Exposures and Immune Dysregulation: Developing Strategies To Improve Malaria Vaccine Efficacy in Young Children. Am J Trop Med Hyg 2024; 110:627-630. [PMID: 38442424 PMCID: PMC10993830 DOI: 10.4269/ajtmh.23-0696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/06/2023] [Indexed: 03/07/2024] Open
Abstract
After several decades in development, two malaria vaccines based on the same antigen and with very similar constructs and adjuvants, RTS,S/AS01 (RTS,S) and R21/Matrix-M (R21), were recommended by the WHO for widespread vaccination of children. These vaccines are much-needed additions to malaria control programs that, when used in conjunction with other control measures, will help to accelerate reductions in malaria morbidity and mortality. Although R21 is not yet available, RTS,S is currently being integrated into routine vaccine schedules in some areas. However, the efficacy of RTS,S is partial, short-lived, and varies widely according to age and geographic location. It is not clear why RTS,S induces protection in some individuals and not others, what the immune mechanisms are that favor protective immunity with RTS,S, and how immune mechanisms are influenced by host and environmental factors. Several studies suggest that higher levels of previous malaria exposure negatively impact RTS,S clinical efficacy. In this article, we summarize data suggesting that previous malaria exposures negatively impact the efficacy of RTS,S and other malaria vaccine candidates. We highlight recent evidence suggesting that increasing malaria exposure impairs the generation of functional antibody responses to RTS,S. Finally, we discuss how investigation of clinical and immune factors associated with suboptimal responses to RTS,S can be used to develop strategies to optimize RTS,S, which will remain relevant to R21 and next-generation vaccines.
Collapse
Affiliation(s)
| | | | - Clarissa Valim
- Boston University School of Public Health, Boston, Massachusetts
| | | |
Collapse
|
19
|
Emmanuel BN, Ishaq AN, Akunne OZ, Saidu UF. Evaluating the knowledge, attitude, perception, and readiness of caregivers of under 5-year-old children to accept malaria vaccine in Nigeria. Clin Exp Vaccine Res 2024; 13:121-131. [PMID: 38752001 PMCID: PMC11091434 DOI: 10.7774/cevr.2024.13.2.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 12/29/2023] [Accepted: 03/28/2024] [Indexed: 05/18/2024] Open
Abstract
Purpose The global burden of disease and mortality is greatly influenced by malaria, particularly in children. Nigeria alone accounts for about 25% of global malaria cases and fatalities. Despite efforts to control and eliminate malaria, conventional treatments have limitations, prompting the need for a vaccine. However, while efforts have focused on researching and developing malaria vaccines, less attention has been given to public acceptance and preparedness for vaccination. Materials and Methods The study employed a cross-sectional approach to assess the knowledge, perceptions, and readiness of caregivers towards the malaria vaccine. Data were collected through a physical and online survey among a representative sample of caregivers across the six geopolitical regions of Nigeria. The data was analyzed using principal component analysis and percentages. Results Out of 347 respondents, 180 (51%) men, 165 (46.6%) women, 2 (0.5%) transgender, 156 (45%) rural settlers, and 191 (55%) urban settlers were identified in this study. The study reported an overall acceptance rate of 78.4% and 21.6% resistance rate. The age group between 21-30 years recorded the highest 207 (59.6%). A significant number of participants, 252 (59.6%), held at least a higher or post-secondary certificate, out of which 193 (55.6%) demonstrated strong readiness to accept the malaria vaccine. The study showed that fear of adverse effects was the main reason for malaria vaccine resistance among caregivers. Conclusion This study's findings offer valuable insights into caregivers' knowledge about the malaria vaccine, highlighting the factors that impact the acceptance of the malaria vaccine.
Collapse
Affiliation(s)
- Blessing Nkechi Emmanuel
- Department of Zoology, Faculty of Life Sciences, Modibbo Adama University of Technology, Yola, Nigeria
| | - Abubakar Nuhu Ishaq
- Department of Biology, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Olisaemeka Zikora Akunne
- Department of Pharmacy, Faculty of Pharmaceutical Science, University of Nigeria Nsukka, Nsukka, Nigeria
| | - Umar Faruk Saidu
- Department of Biochemistry and Molecular Biology, Faculty of Life Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
| |
Collapse
|
20
|
Goswami D, Patel H, Betz W, Armstrong J, Camargo N, Patil A, Chakravarty S, Murphy SC, Sim BKL, Vaughan AM, Hoffman SL, Kappe SH. A replication competent Plasmodium falciparum parasite completely attenuated by dual gene deletion. EMBO Mol Med 2024; 16:723-754. [PMID: 38514791 PMCID: PMC11018819 DOI: 10.1038/s44321-024-00057-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024] Open
Abstract
Vaccination with infectious Plasmodium falciparum (Pf) sporozoites (SPZ) administered with antimalarial drugs (PfSPZ-CVac), confers superior sterilizing protection against infection when compared to vaccination with replication-deficient, radiation-attenuated PfSPZ. However, the requirement for drug administration constitutes a major limitation for PfSPZ-CVac. To obviate this limitation, we generated late liver stage-arresting replication competent (LARC) parasites by deletion of the Mei2 and LINUP genes (mei2-/linup- or LARC2). We show that Plasmodium yoelii (Py) LARC2 sporozoites did not cause breakthrough blood stage infections and engendered durable sterilizing immunity against various infectious sporozoite challenges in diverse strains of mice. We next genetically engineered a PfLARC2 parasite strain that was devoid of extraneous DNA and produced cryopreserved PfSPZ-LARC2. PfSPZ-LARC2 liver stages replicated robustly in liver-humanized mice but displayed severe defects in late liver stage differentiation and did not form liver stage merozoites. This resulted in complete abrogation of parasite transition to viable blood stage infection. Therefore, PfSPZ-LARC2 is the next-generation vaccine strain expected to unite the safety profile of radiation-attenuated PfSPZ with the superior protective efficacy of PfSPZ-CVac.
Collapse
Affiliation(s)
- Debashree Goswami
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Hardik Patel
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - William Betz
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Janna Armstrong
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Nelly Camargo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Asha Patil
- Sanaria Inc., 9800 Medical Center Dr., Rockville, MD, 20850, USA
| | | | - Sean C Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - B Kim Lee Sim
- Sanaria Inc., 9800 Medical Center Dr., Rockville, MD, 20850, USA
| | - Ashley M Vaughan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Stefan Hi Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
21
|
Locke E, Flores-Garcia Y, Mayer BT, MacGill RS, Borate B, Salgado-Jimenez B, Gerber MW, Mathis-Torres S, Shapiro S, King CR, Zavala F. Establishing RTS,S/AS01 as a benchmark for comparison to next-generation malaria vaccines in a mouse model. NPJ Vaccines 2024; 9:29. [PMID: 38341502 DOI: 10.1038/s41541-024-00819-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
New strategies are needed to reduce the incidence of malaria, and promising approaches include vaccines targeting the circumsporozoite protein (CSP). To improve upon the malaria vaccine, RTS,S/AS01, it is essential to standardize preclinical assays to measure the potency of next-generation vaccines against this benchmark. We focus on RTS,S/AS01-induced antibody responses and functional activity in conjunction with robust statistical analyses. Transgenic Plasmodium berghei sporozoites containing full-length P. falciparum CSP (tgPb-PfCSP) allow two assessments of efficacy: quantitative reduction in liver infection following intravenous challenge, and sterile protection from mosquito bite challenge. Two or three doses of RTS,S/AS01 were given intramuscularly at 3-week intervals, with challenge 2-weeks after the last vaccination. Minimal inter- and intra-assay variability indicates the reproducibility of the methods. Importantly, the range of this model is suitable for screening more potent vaccines. Levels of induced anti-CSP antibody 2A10 equivalency were also associated with activity: 105 μg/mL (95% CI: 68.8, 141) reduced liver infection by 50%, whereas 285 μg/mL (95% CI: 166, 404) is required for 50% sterile protection from mosquito bite challenge. Additionally, the liver burden model was able to differentiate between protected and non-protected human plasma samples from a controlled human malaria infection study, supporting these models' relevance and predictive capability. Comparison in animal models of CSP-based vaccine candidates to RTS,S/AS01 is now possible under well controlled conditions. Assessment of the quality of induced antibodies, likely a determinant of durability of protection in humans, should be possible using these methods.
Collapse
Affiliation(s)
- Emily Locke
- Center for Vaccine Innovation and Access, PATH, Washington, DC, 20001, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Bryan T Mayer
- Vaccine and Infectious Disease Division, Fred Hutchison Cancer Research Center, Seattle, WA, 98109, USA
| | - Randall S MacGill
- Center for Vaccine Innovation and Access, PATH, Washington, DC, 20001, USA
| | - Bhavesh Borate
- Vaccine and Infectious Disease Division, Fred Hutchison Cancer Research Center, Seattle, WA, 98109, USA
| | - Berenice Salgado-Jimenez
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Monica W Gerber
- Vaccine and Infectious Disease Division, Fred Hutchison Cancer Research Center, Seattle, WA, 98109, USA
| | - Shamika Mathis-Torres
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sarah Shapiro
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - C Richter King
- Center for Vaccine Innovation and Access, PATH, Washington, DC, 20001, USA
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
22
|
MacMillen Z, Hatzakis K, Simpson A, Shears MJ, Watson F, Erasmus JH, Khandhar AP, Wilder B, Murphy SC, Reed SG, Davie JW, Avril M. Accelerated prime-and-trap vaccine regimen in mice using repRNA-based CSP malaria vaccine. NPJ Vaccines 2024; 9:12. [PMID: 38200025 PMCID: PMC10781674 DOI: 10.1038/s41541-023-00799-4] [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: 06/09/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Malaria, caused by Plasmodium parasites, remains one of the most devastating infectious diseases worldwide, despite control efforts to lower morbidity and mortality. Both advanced candidate vaccines, RTS,S and R21, are subunit (SU) vaccines that target a single Plasmodium falciparum (Pf) pre-erythrocytic (PE) sporozoite (spz) surface protein known as circumsporozoite (CS). These vaccines induce humoral immunity but fail to elicit CD8 + T-cell responses sufficient for long-term protection. In contrast, whole-organism (WO) vaccines, such as Radiation Attenuated Sporozoites (RAS), achieved sterile protection but require a series of intravenous doses administered in multiple clinic visits. Moreover, these WO vaccines must be produced in mosquitos, a burdensome process that severely limits their availability. To reduce reliance on WO while maintaining protection via both antibodies and Trm responses, we have developed an accelerated vaccination regimen that combines two distinct agents in a prime-and-trap strategy. The priming dose is a single dose of self-replicating RNA encoding the full-length P. yoelii CS protein, delivered via an advanced cationic nanocarrier (LIONTM). The trapping dose consists of one dose of WO RAS. Our vaccine induces a strong immune response when administered in an accelerated regimen, i.e., either 5-day or same-day immunization. Additionally, mice after same-day immunization showed a 2-day delay of blood patency with 90% sterile protection against a 3-week spz challenge. The same-day regimen also induced durable 70% sterile protection against a 2-month spz challenge. Our approach presents a clear path to late-stage preclinical and clinical testing of dose-sparing, same-day regimens that can confer sterilizing protection against malaria.
Collapse
Affiliation(s)
- Zachary MacMillen
- MalarVx, Inc 1551 Eastlake Ave E, Suite 100, Seattle, WA, 98102, USA
| | - Kiara Hatzakis
- MalarVx, Inc 1551 Eastlake Ave E, Suite 100, Seattle, WA, 98102, USA
| | - Adrian Simpson
- HDT Bio, 1150 Eastlake Ave E, Suite 200A, Seattle, WA, 98109, USA
| | - Melanie J Shears
- University of Washington, Department of Laboratory Medicine and Pathology, 750 Republican St., F870, Seattle, WA, 98109, USA
| | - Felicia Watson
- University of Washington, Department of Laboratory Medicine and Pathology, 750 Republican St., F870, Seattle, WA, 98109, USA
| | - Jesse H Erasmus
- HDT Bio, 1150 Eastlake Ave E, Suite 200A, Seattle, WA, 98109, USA
| | - Amit P Khandhar
- HDT Bio, 1150 Eastlake Ave E, Suite 200A, Seattle, WA, 98109, USA
| | - Brandon Wilder
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Building 1, Room 2220, 505 NW 185th Ave, Beaverton, OR, 97006, USA
| | - Sean C Murphy
- University of Washington, Department of Laboratory Medicine and Pathology, 750 Republican St., F870, Seattle, WA, 98109, USA
| | - Steven G Reed
- HDT Bio, 1150 Eastlake Ave E, Suite 200A, Seattle, WA, 98109, USA
| | - James W Davie
- MalarVx, Inc 1551 Eastlake Ave E, Suite 100, Seattle, WA, 98102, USA
| | - Marion Avril
- MalarVx, Inc 1551 Eastlake Ave E, Suite 100, Seattle, WA, 98102, USA.
| |
Collapse
|
23
|
Mura M, Misganaw B, Gautam A, Robinson T, Chaudhury S, Bansal N, Martins AJ, Tsang J, Hammamieh R, Bergmann-Leitner E. Human transcriptional signature of protection after Plasmodium falciparum immunization and infectious challenge via mosquito bites. Hum Vaccin Immunother 2023; 19:2282693. [PMID: 38010150 PMCID: PMC10760396 DOI: 10.1080/21645515.2023.2282693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/08/2023] [Indexed: 11/29/2023] Open
Abstract
The identification of immune correlates of protection against infectious pathogens will accelerate the design and optimization of recombinant and subunit vaccines. Systematic analyses such as immunoprofiling including serological, cellular, and molecular assessments supported by computational tools are key to not only identify correlates of protection but also biomarkers of disease susceptibility. The current study expands our previous cellular and serological profiling of vaccine-induced responses to a whole parasite malaria vaccine. The irradiated sporozoite model was chosen as it is considered the most effective vaccine against malaria. In contrast to whole blood transcriptomics analysis, we stimulated peripheral blood mononuclear cells (PBMC) with sporozoites and enriched for antigen-specific cells prior to conducting transcriptomics analysis. By focusing on transcriptional events triggered by antigen-specific stimulation, we were able to uncover quantitative and qualitative differences between protected and non-protected individuals to controlled human malaria infections and identified differentially expressed genes associated with sporozoite-specific responses. Further analyses including pathway and gene set enrichment analysis revealed that vaccination with irradiated sporozoites induced a transcriptomic profile associated with Th1-responses, Interferon-signaling, antigen-presentation, and inflammation. Analyzing longitudinal time points not only post-vaccination but also post-controlled human malaria infection further revealed that the transcriptomic profile of protected vs non-protected individuals was not static but continued to diverge over time. The results lay the foundation for comparing protective immune signatures induced by various vaccine platforms to uncover immune correlates of protection that are common across platforms.
Collapse
Affiliation(s)
- Marie Mura
- Immunology Core, Biologics Research & Development, WRAIR-Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Host-Pathogen Interactions, Microbiology and Infectious Diseases, IRBA-Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Burook Misganaw
- Medical Readiness Systems Biology, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Vysnova Inc, Landover, MD, USA
| | - Aarti Gautam
- Medical Readiness Systems Biology, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Tanisha Robinson
- Immunology Core, Biologics Research & Development, WRAIR-Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Sidhartha Chaudhury
- Center of Enabling Capabilties, WRAIR-Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Neha Bansal
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Andrew J. Martins
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - John Tsang
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | - Rasha Hammamieh
- Medical Readiness Systems Biology, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Elke Bergmann-Leitner
- Immunology Core, Biologics Research & Development, WRAIR-Walter Reed Army Institute of Research, Silver Spring, MD, USA
| |
Collapse
|
24
|
Amin MA, Afrin S, Bonna AS, Rozars MFK, Nabi MH, Hawlader MDH. Knowledge and acceptance of malaria vaccine among parents of under-five children of malaria endemic areas in Bangladesh: A cross-sectional study. Health Expect 2023; 26:2630-2643. [PMID: 37661603 PMCID: PMC10632622 DOI: 10.1111/hex.13862] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/05/2023] Open
Abstract
BACKGROUND Malaria exists as an endemic in many countries including Bangladesh and the malaria vaccine is not yet available here. The study aimed to assess the level of knowledge and acceptance of the malaria vaccination among the parents of children under the age of five in Bangladesh's malaria-endemic areas and the sociodemographic, behavioural, and household factors associated with the acceptance and knowledge of the malaria vaccine. METHODS From January to March 2022, a cross-sectional study was conducted in all five malaria-endemic districts of Bangladesh, involving 405 parents of children under the age of 5 who met the inclusion criteria. Multiple logistic regression was used to analyze the factor affecting parents' acceptance and knowledge of malaria vaccination in children under five and other variables. RESULTS Majority (54%) of the respondents were mothers. Almost half (49%) of the respondents were aged between 26 and 35 years old and around 90% were from rural areas. A small portion (20%) of the participants were housewives and 46% of them completed primary education. Overall, 70% of the study participants reported that they would accept malaria vaccination independently. About one-fourth (25%) heard about the malaria vaccine and 48% of them mentioned health professionals as the source of information. Knowledge of malaria vaccination was found associated with residence, income, and family size. Acceptance and knowledge were both associated with residence, education, occupation, income, and family size. In a multivariable analysis, housing structure, house wall, house window, knowledge of malaria, testing for malaria, and being diagnosed with malaria were all associated with knowledge of and acceptance of getting vaccinated against malaria. CONCLUSIONS The present study highlights the necessity of creating awareness of malaria vaccines in epidemic areas of Bangladesh. This study offers crucial data to develop a policy for a novel malaria vaccine, supporting its adoption in Bangladesh. PUBLIC CONTRIBUTION This study was based on interviews. The interviewees were recruited as public representatives from the malaria-endemic area to assist us in building an understanding of knowledge and acceptance of the malaria vaccine among parents of under-five children in Bangladesh.
Collapse
Affiliation(s)
- Mohammad Ashraful Amin
- Department of Public HealthNorth South UniversityDhakaBangladesh
- Public Health Professional Development Society (PPDS)DhakaBangladesh
| | - Sadia Afrin
- Department of Public HealthNorth South UniversityDhakaBangladesh
- Public Health Professional Development Society (PPDS)DhakaBangladesh
| | - Atia S. Bonna
- Department of Public HealthNorth South UniversityDhakaBangladesh
- Public Health Professional Development Society (PPDS)DhakaBangladesh
- Public Health Epidemiologist, HN & HIV SectorSave the ChildrenDhakaBangladesh
| | - Md Faisal K. Rozars
- Department of Public HealthNorth South UniversityDhakaBangladesh
- Public Health Professional Development Society (PPDS)DhakaBangladesh
| | | | | |
Collapse
|
25
|
Hoyt J, Okello G, Bange T, Kariuki S, Jalloh MF, Webster J, Hill J. RTS,S/AS01 malaria vaccine pilot implementation in western Kenya: a qualitative longitudinal study to understand immunisation barriers and optimise uptake. BMC Public Health 2023; 23:2283. [PMID: 37980467 PMCID: PMC10657022 DOI: 10.1186/s12889-023-17194-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/09/2023] [Indexed: 11/20/2023] Open
Abstract
BACKGROUND Malaria is a significant public health threat in sub-Saharan Africa, particularly among children. The RTS,S/AS01 malaria vaccine reduces the risk and severity of malaria in children. RTS,S/AS01 was piloted in three African countries, Ghana, Kenya and Malawi, to assess safety, feasibility and cost-effectiveness in real-world settings. A qualitative longitudinal study was conducted as part of the feasibility assessment. This analysis explores RTS,S/AS01 vaccination barriers and identifies potential motivators among caregivers in three sub-counties in western Kenya. METHODS A cohort of 63 caregivers with a malaria vaccine eligible child was interviewed at three time points over 24 months. A sub-set of 11 caregivers whose eligible children were either partially or non-vaccinated were selected for this sub-analysis. The 5A Taxonomy for root causes of under-vaccination was used to organise the inductively-coded data into categories (awareness, acceptance, access, affordability, and activation) and identify the factors influencing uptake across caregivers. A trajectory analysis was conducted to understand changes in factors over time within each caregiver experience. Caregiver narratives are used to illustrate how the factors influencing uptake were interrelated and changed over time. RESULTS Lack of awareness, previous negative experiences with routine childhood immunisations and the burden of getting to the health facility contributed to caregivers initially delaying uptake of the vaccine. Over time concerns about vaccine side effects diminished and anticipated vaccination benefits strongly motivated caregivers to vaccinate their children. Persistent health system barriers (e.g., healthcare provider strikes, vaccine stockouts, negative provider attitudes) meant some children missed the first-dose eligibility window by aging-out. CONCLUSIONS Caregivers in this study believed the RTS,S/AS01 to be effective and were motivated to have their children vaccinated. Despite these positive perceptions of the malaria vaccine, uptake was substantially hindered by persistent health system constraints. Negative provider attitudes emerged as a powerful deterrent to attending immunisation services and hampered uptake of the vaccine. Strategies that focus on improving interpersonal communication skills among healthcare providers are needed.
Collapse
Affiliation(s)
- Jenna Hoyt
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
| | - George Okello
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
- Kenya Medical Research Institute/Centre for Global Health Research, Kisumu, Kenya
| | - Teresa Bange
- Kenya Medical Research Institute/Centre for Global Health Research, Kisumu, Kenya
| | - Simon Kariuki
- Kenya Medical Research Institute/Centre for Global Health Research, Kisumu, Kenya
| | - Mohamed F Jalloh
- Global Immunization Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jayne Webster
- Disease Control Department, London School of Tropical Medicine and Hygiene, London, UK
| | - Jenny Hill
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
26
|
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
|
27
|
Ohene-Adjei K, Asante KP, Akuffo KO, Tounaikok N, Asiamah M, Owiredu D, Manu AA, Danso-Appiah A. Malaria vaccine-related adverse events among children under 5 in sub-Saharan Africa: systematic review and meta-analysis protocol. BMJ Open 2023; 13:e076985. [PMID: 37793915 PMCID: PMC10551995 DOI: 10.1136/bmjopen-2023-076985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023] Open
Abstract
INTRODUCTION The RTS,S vaccine has been approved for use in children under 5 living in moderate to high malaria transmission areas. However, clinically important adverse events have been reported in countries in sub-Saharan Africa. This systematic review aims to assess the frequency, severity and clinical importance of vaccine-related adverse events. METHODS AND ANALYSIS This systematic review protocol has been prepared following robust methods and reported in line with the Preferred Reporting Items for Systematic reviews and Meta-Analyses for protocols guidelines. We will search PubMed, CINAHL, LILACS, Google Scholar, SCOPUS, WEB OF SCIENCE, Cochrane library, HINARI, African Journals Online, Trip Pro and TOXNET from 2000 to 30 September 2023, without language restrictions. We will also search conference proceedings, dissertations, World Bank Open Knowledge Repository, and WHO, PATH, UNICEF, Food and Drugs Authorities and European Medicines Agency databases, preprint repositories and reference lists of relevant studies for additional studies. Experts in the field will be contacted for unpublished or published studies missed by our searches. At least two reviewers will independently select studies and extract data using pretested tools and assess risk of bias in the included studies using the Cochrane risk of bias tool. Any disagreements will be resolved through discussion between the reviewers. Heterogeneity will be explored graphically, and statistically using the I2 statistic. We will conduct random-effects meta-analysis when heterogeneity is appreciable, and express dichotomous outcomes (serious adverse events, cerebral malaria and febrile convulsion) as risk ratio (RR) with their 95% CI. We will perform subgroup analysis to assess the impact of heterogeneity and sensitivity analyses to test the robustness of the effect estimates. The overall level of evidence will be assessed using Grading of Recommendations Assessment, Development and Evaluation. ETHICS AND DISSEMINATION Ethical approval is not required for a systematic review. The findings of this study will be disseminated through stakeholder forums, conferences and peer-review publications. PROSPERO REGISTRATION NUMBER CRD42021275155.
Collapse
Affiliation(s)
- Kennedy Ohene-Adjei
- Department of Epidemiology and Disease Control, School of Public Health, College of Health Sciences, University of Ghana, Accra, Ghana
- Tain District Health Directorate, Ghana Health Service, Tain, Ghana
| | - Kwaku Poku Asante
- Research and Development Division, Kintampo Health Research Centre, Ghana Health Service, Kintampo, Kintampo North Municipality, Bono East Region, Ghana
| | - Kwadwo Owusu Akuffo
- Department of Optometry and Visual Science, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Narcisse Tounaikok
- Centre for Evidence Synthesis and Policy, School of Public Health, University of Ghana, Accra, Ghana
- Department of Human and Animal Health, University of Emi Koussi, N'Djamena, Chad
| | - Morrison Asiamah
- Department of Electron Microscopy and Histopathology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - David Owiredu
- Department of Epidemiology and Disease Control, School of Public Health, College of Health Sciences, University of Ghana, Accra, Ghana
- Centre for Evidence Synthesis and Policy, School of Public Health, University of Ghana, Accra, Ghana
| | - Alexander Ansah Manu
- Department of Epidemiology and Disease Control, School of Public Health, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Anthony Danso-Appiah
- Department of Epidemiology and Disease Control, School of Public Health, College of Health Sciences, University of Ghana, Accra, Ghana
- Centre for Evidence Synthesis and Policy, School of Public Health, University of Ghana, Accra, Ghana
| |
Collapse
|
28
|
Nakamae S, Miyagawa S, Ogawa K, Kamiya M, Taniguchi M, Ono A, Kawaguchi M, Teklemichael AA, Jian JY, Araki T, Katagami Y, Mukai H, Annoura T, Yui K, Hirayama K, Kawakami S, Mizukami S. Induction of liver-resident memory T cells and protection at liver-stage malaria by mRNA-containing lipid nanoparticles. Front Immunol 2023; 14:1116299. [PMID: 37680630 PMCID: PMC10482405 DOI: 10.3389/fimmu.2023.1116299] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 07/25/2023] [Indexed: 09/09/2023] Open
Abstract
Recent studies have suggested that CD8+ liver-resident memory T (TRM) cells are crucial in the protection against liver-stage malaria. We used liver-directed mRNA-containing lipid nanoparticles (mRNA-LNPs) to induce liver TRM cells in a murine model. Single-dose intravenous injections of ovalbumin mRNA-LNPs effectively induced antigen-specific cytotoxic T lymphocytes in a dose-dependent manner in the liver on day 7. TRM cells (CD8+ CD44hi CD62Llo CD69+ KLRG1-) were induced 5 weeks after immunization. To examine the protective efficacy, mice were intramuscularly immunized with two doses of circumsporozoite protein mRNA-LNPs at 3-week intervals and challenged with sporozoites of Plasmodium berghei ANKA. Sterile immunity was observed in some of the mice, and the other mice showed a delay in blood-stage development when compared with the control mice. mRNA-LNPs therefore induce memory CD8+ T cells that can protect against sporozoites during liver-stage malaria and may provide a basis for vaccines against the disease.
Collapse
Affiliation(s)
- Sayuri Nakamae
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Satoshi Miyagawa
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
- Laboratory for Drug Discovery and Disease Research, SHIONOGI & CO., LTD., Osaka, Japan
| | - Koki Ogawa
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Mariko Kamiya
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Mayumi Taniguchi
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Akari Ono
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Maho Kawaguchi
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Awet Alem Teklemichael
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Jiun-Yu Jian
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Tamasa Araki
- Department of Parasitology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Yukimi Katagami
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Hidefumi Mukai
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Takeshi Annoura
- Department of Parasitology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Katsuyuki Yui
- Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Kenji Hirayama
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Shusaku Mizukami
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Nagasaki, Japan
| |
Collapse
|
29
|
Genito CJ, Brooks K, Smith A, Ryan E, Soto K, Li Y, Warter L, Dutta S. Protective antibody threshold of RTS,S/AS01 malaria vaccine correlates antigen and adjuvant dose in mouse model. NPJ Vaccines 2023; 8:114. [PMID: 37563255 PMCID: PMC10415390 DOI: 10.1038/s41541-023-00714-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023] Open
Abstract
Mouse models are useful for the early down-selection of malaria vaccine candidates. The Walter Reed Army Institute of Research has optimized a transgenic Plasmodium berghei sporozoite challenge model to compare the efficacy of Plasmodium falciparum circumsporozoite protein (CSP) vaccines. GSK's RTS,S vaccine formulated in the adjuvant AS01 can protect malaria-naïve individuals against malaria. We report that the RTS,S/AS01 vaccine induces high level sterile protection in our mouse model. Down titration of the antigen at a constant AS01 dose revealed a potent antigen dose-sparing effect and the superiority of RTS,S/AS01 over a soluble CSP antigen. RTS,S-mediated protective immunity was associated with a threshold of major repeat antibody titer. Combined titration of the antigen and adjuvant showed that reducing the adjuvant could improve antibody boosting post-3rd vaccination and reduce the threshold antibody concentration required for protection. Mouse models can provide a pathway for preclinical assessment of strategies to improve CSP vaccines against malaria.
Collapse
Affiliation(s)
- Christopher J Genito
- Structural Vaccinology Laboratory, Biologics Research and Development Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Katherine Brooks
- Structural Vaccinology Laboratory, Biologics Research and Development Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Alexis Smith
- Structural Vaccinology Laboratory, Biologics Research and Development Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Emma Ryan
- Structural Vaccinology Laboratory, Biologics Research and Development Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Kim Soto
- Structural Vaccinology Laboratory, Biologics Research and Development Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Yuanzhang Li
- Center for Enabling Capabilities, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | | | - Sheetij Dutta
- Structural Vaccinology Laboratory, Biologics Research and Development Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA.
| |
Collapse
|
30
|
Nikas A, Ahmed H, Zarnitsyna VI. Competing Heterogeneities in Vaccine Effectiveness Estimation. Vaccines (Basel) 2023; 11:1312. [PMID: 37631880 PMCID: PMC10458793 DOI: 10.3390/vaccines11081312] [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: 06/20/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Understanding the waning of vaccine-induced protection is important for both immunology and public health. Population heterogeneities in underlying (pre-vaccination) susceptibility and vaccine response can cause measured vaccine effectiveness (mVE) to change over time, even in the absence of pathogen evolution and any actual waning of immune responses. We use multi-scale agent-based models parameterized using epidemiological and immunological data, to investigate the effect of these heterogeneities on mVE as measured by the hazard ratio. Based on our previous work, we consider the waning of antibodies according to a power law and link it to protection in two ways: (1) motivated by correlates of risk data and (2) using a within-host model of stochastic viral extinction. The effect of the heterogeneities is given by concise and understandable formulas, one of which is essentially a generalization of Fisher's fundamental theorem of natural selection to include higher derivatives. Heterogeneity in underlying susceptibility accelerates apparent waning, whereas heterogeneity in vaccine response slows down apparent waning. Our models suggest that heterogeneity in underlying susceptibility is likely to dominate. However, heterogeneity in vaccine response offsets <10% to >100% (median of 29%) of this effect in our simulations. Our study suggests heterogeneity is more likely to 'bias' mVE downwards towards the faster waning of immunity but a subtle bias in the opposite direction is also plausible.
Collapse
Affiliation(s)
- Ariel Nikas
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hasan Ahmed
- Department of Biology, Emory University, Atlanta, GA 30322, USA;
| | - Veronika I. Zarnitsyna
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
31
|
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
|
32
|
MacMillen Z, Hatzakis K, Simpson A, Shears M, Watson F, Erasmus J, Khandhar A, Wilder B, Murphy S, Reed S, Davie J, Avril M. Accelerated prime-and-trap vaccine regimen in mice using repRNA-based CSP malaria vaccine. RESEARCH SQUARE 2023:rs.3.rs-3045076. [PMID: 37461621 PMCID: PMC10350175 DOI: 10.21203/rs.3.rs-3045076/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
Malaria, caused by Plasmodium parasites, remains one of the most devastating infectious diseases worldwide, despite control efforts that have lowered morbidity and mortality. The only P. falciparum vaccine candidates to show field efficacy are those targeting the asymptomatic pre-erythrocytic (PE) stages of infection. The subunit (SU) RTS,S/AS01 vaccine, the only licensed malaria vaccine to date, is only modestly effective against clinical malaria. Both RTS,S/AS01 and the SU R21 vaccine candidate target the PE sporozoite (spz) circumsporozoite (CS) protein. These candidates elicit high-titer antibodies that provide short-term protection from disease, but do not induce the liver-resident memory CD8+ T cells (Trm) that confer strong PE immunity and long-term protection. In contrast, whole-organism (WO) vaccines, employing for example radiation-attenuated spz (RAS), elicit both high antibody titers and Trm, and have achieved high levels of sterilizing protection. However, they require multiple intravenous (IV) doses, which must be administered at intervals of several weeks, complicating mass administration in the field. Moreover, the quantities of spz required present production difficulties. To reduce reliance on WO while maintaining protection via both antibodies and Trm responses, we have developed an accelerated vaccination regimen that combines two distinct agents in a prime-and-trap strategy. While the priming dose is a self-replicating RNA encoding P. yoelii CS protein, delivered via an advanced cationic nanocarrier (LION™), the trapping dose consists of WO RAS. This accelerated regime confers sterile protection in the P. yoelii mouse model of malaria. Our approach presents a clear path to late-stage preclinical and clinical testing of dose-sparing, same-day regimens that can confer sterilizing protection against malaria.
Collapse
|
33
|
Chuang YM, Alameh MG, Abouneameh S, Raduwan H, Ledizet M, Weissman D, Fikrig E. A mosquito AgTRIO mRNA vaccine contributes to immunity against malaria. NPJ Vaccines 2023; 8:88. [PMID: 37286568 PMCID: PMC10244833 DOI: 10.1038/s41541-023-00679-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/16/2023] [Indexed: 06/09/2023] Open
Abstract
Malaria begins when an infected mosquito injects saliva containing Plasmodium sporozoites into the skin of a vertebrate host. To prevent malaria, vaccination is the most effective strategy and there is an urgent need for new strategies to enhance current pathogen-based vaccines. Active or passive immunization against a mosquito saliva protein, AgTRIO, contributes to protection against Plasmodium infection of mice. In this study, we generated an AgTRIO mRNA-lipid nanoparticle (LNP) and assessed its potential usefulness as a vaccine against malaria. Immunization of mice with an AgTRIO mRNA-LNP generated a robust humoral response, including AgTRIO IgG2a isotype antibodies that have been associated with protection. AgTRIO mRNA-LNP immunized mice exposed to Plasmodium berghei-infected mosquitoes had markedly reduced initial Plasmodium hepatic infection levels and increased survival compared to control mice. In addition, as the humoral response to AgTRIO waned over 6 months, additional mosquito bites boosted the AgTRIO IgG titers, including IgG1 and IgG2a isotypes, which offers a unique advantage compared to pathogen-based vaccines. These data will aid in the generation of future malaria vaccines that may include both pathogen and vector antigens.
Collapse
Affiliation(s)
- Yu-Min Chuang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | - Mohamad-Gabriel Alameh
- Institute for RNA Innovation and Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Selma Abouneameh
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Hamidah Raduwan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | - Drew Weissman
- Institute for RNA Innovation and Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
34
|
MacMillen Z, Hatzakis K, Simpson A, Shears MJ, Watson F, Erasmus JH, Khandhar AP, Wilder B, Murphy SC, Reed SG, Davie JW, Avril M. Accelerated prime-and-trap vaccine regimen in mice using repRNA-based CSP malaria vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.23.541932. [PMID: 37292739 PMCID: PMC10245832 DOI: 10.1101/2023.05.23.541932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Malaria, caused by Plasmodium parasites, remains one of the most devastating infectious diseases worldwide, despite control efforts that have lowered morbidity and mortality. The only P. falciparum vaccine candidates to show field efficacy are those targeting the asymptomatic pre-erythrocytic (PE) stages of infection. The subunit (SU) RTS,S/AS01 vaccine, the only licensed malaria vaccine to date, is only modestly effective against clinical malaria. Both RTS,S/AS01 and the SU R21 vaccine candidate target the PE sporozoite (spz) circumsporozoite (CS) protein. These candidates elicit high-titer antibodies that provide short-term protection from disease, but do not induce the liver-resident memory CD8+ T cells (Trm) that confer strong PE immunity and long-term protection. In contrast, whole-organism (WO) vaccines, employing for example radiation-attenuated spz (RAS), elicit both high antibody titers and Trm, and have achieved high levels of sterilizing protection. However, they require multiple intravenous (IV) doses, which must be administered at intervals of several weeks, complicating mass administration in the field. Moreover, the quantities of spz required present production difficulties. To reduce reliance on WO while maintaining protection via both antibodies and Trm responses, we have developed an accelerated vaccination regimen that combines two distinct agents in a prime-and-trap strategy. While the priming dose is a self-replicating RNA encoding P. yoelii CS protein, delivered via an advanced cationic nanocarrier (LION™), the trapping dose consists of WO RAS. This accelerated regime confers sterile protection in the P. yoelii mouse model of malaria. Our approach presents a clear path to late-stage preclinical and clinical testing of dose-sparing, same-day regimens that can confer sterilizing protection against malaria.
Collapse
Affiliation(s)
| | - Kiara Hatzakis
- MalarVx, Inc 1551 Eastlake Ave E, Suite 100, Seattle WA 98102
| | - Adrian Simpson
- HDT Bio, 1616 Eastlake Ave E, Suite 280, Seattle WA 98102
| | - Melanie J. Shears
- University of Washington, Department of Laboratory Medicine and Pathology, 750 Republican St., F870, Seattle, WA 98109
| | - Felicia Watson
- University of Washington, Department of Laboratory Medicine and Pathology, 750 Republican St., F870, Seattle, WA 98109
| | | | | | - Brandon Wilder
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Building 1, Room 2220, 505 NW 185th Ave, Beaverton, OR 97006
| | - Sean C. Murphy
- University of Washington, Department of Laboratory Medicine and Pathology, 750 Republican St., F870, Seattle, WA 98109
| | - Steven G. Reed
- HDT Bio, 1616 Eastlake Ave E, Suite 280, Seattle WA 98102
| | - James W. Davie
- MalarVx, Inc 1551 Eastlake Ave E, Suite 100, Seattle WA 98102
| | - Marion Avril
- MalarVx, Inc 1551 Eastlake Ave E, Suite 100, Seattle WA 98102
| |
Collapse
|
35
|
El-Moamly AA, El-Sweify MA. Malaria vaccines: the 60-year journey of hope and final success-lessons learned and future prospects. Trop Med Health 2023; 51:29. [PMID: 37198702 DOI: 10.1186/s41182-023-00516-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/18/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND The world has made great strides towards beating malaria, although about half of the world population is still exposed to the risk of contracting malaria. Developing an effective malaria vaccine was a huge challenge for medical science. In 2021 the World Health Organization (WHO) approved the first malaria vaccine, RTS,S/AS01 vaccine (Mosquirix™), for widespread use. This review highlights the history of development, and the different approaches and types of malaria vaccines, and the literature to date. It covers the developmental stages of RTS,S/AS01 and recommends steps for its deployment. The review explores other potential vaccine candidates and their status, and suggests options for their further development. It also recommends future roles for vaccines in eradicating malaria. Questions remain on how RTS,S vaccine will work in widespread use and how it can best be utilized to benefit vulnerable communities. CONCLUSION Malaria vaccines have been in development for almost 60 years. The RTS,S/AS01 vaccine has now been approved, but cannot be a stand-alone solution. Development should continue on promising candidates such as R21, PfSPZ and P. vivax vaccines. Multi-component vaccines may be a useful addition to other malaria control techniques in achieving eradication of malaria.
Collapse
Affiliation(s)
- Amal A El-Moamly
- Department of Medical Parasitology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Mohamed A El-Sweify
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
36
|
Gbedande K, Ibitokou SA, Ong ML, Degli-Esposti MA, Brown MG, Stephens R. Boosting Live Malaria Vaccine with Cytomegalovirus Vector Can Prolong Immunity through Innate and Adaptive Mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.02.539025. [PMID: 37205446 PMCID: PMC10187235 DOI: 10.1101/2023.05.02.539025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Vaccines to persistent parasite infections have been challenging, and current iterations lack long-term protection. Cytomegalovirus (CMV) chronic vaccine vectors drive protection against SIV, tuberculosis and liver-stage malaria correlated with antigen-specific CD8 T cells with a Tem phenotype. This phenotype is likely driven by a combination of antigen-specific and innate adjuvanting effects of the vector, though these mechanisms are less well understood. Sterilizing immunity from live Plasmodium chabaudi vaccination lasts less than 200 days. While P. chabaudi-specific antibody levels remain stable after vaccination, the decay of parasite-specific T cells correlates with loss of challenge protection. Therefore, we enlisted murine CMV as a booster strategy to prolong T cell responses against malaria. To study induced T cell responses, we included P. chabaudi MSP-1 epitope B5 (MCMV-B5). We found that MCMV vector alone significantly protected against a challenge P. chabaudi infection 40-60 days later, and that MCMV-B5 was able to make B5-specific Teff, in addition to previously-reported Tem, that survive to the challenge timepoint. Used as a booster, MCMV-B5 prolonged protection from heterologous infection beyond day 200, and increased B5 TCR Tg T cell numbers, including both a highly-differentiated Tem phenotype and Teff, both previously reported to protect. B5 epitope expression was responsible for maintenance of Th1 and Tfh B5 T cells. In addition, the MCMV vector had adjuvant properties, contributing non-specifically through prolonged stimulation of IFN-γ. In vivo neutralization of IFN-γ, but not IL-12 and IL-18, late in the course of MCMV, led to loss of the adjuvant effect. Mechanistically, sustained IFN-γ from MCMV increased CD8α+ dendritic cell numbers, and led to increased IL-12 production upon Plasmodium challenge. In addition, neutralization of IFN-γ before challenge reduced the polyclonal Teff response to challenge. Our findings suggest that, as protective epitopes are defined, an MCMV vectored booster can prolong protection through the innate effects of IFN-γ.
Collapse
Affiliation(s)
- Komi Gbedande
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555-0435
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Cancer Center, 205 S. Orange Avenue, Newark, NJ 07103
| | - Samad A Ibitokou
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555-0435
| | - Monique L Ong
- Centre for Experimental Immunology, Lions Eye Institute; Nedlands, Western Australia, Australia
- Infection and Immunity Program and Department of Microbiology, Biomedicine Discovery Institute, Monash University; Clayton, Victoria, Australia
| | - Mariapia A Degli-Esposti
- Centre for Experimental Immunology, Lions Eye Institute; Nedlands, Western Australia, Australia
- Infection and Immunity Program and Department of Microbiology, Biomedicine Discovery Institute, Monash University; Clayton, Victoria, Australia
| | - Michael G Brown
- Department of Medicine, Division of Nephrology, and the Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA
| | - Robin Stephens
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555-0435
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Cancer Center, 205 S. Orange Avenue, Newark, NJ 07103
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
37
|
Nikas A, Ahmed H, Zarnitsyna VI. Competing Heterogeneities in Vaccine Effectiveness Estimation. ARXIV 2023:arXiv:2305.01737v2. [PMID: 37205263 PMCID: PMC10187365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Understanding waning of vaccine-induced protection is important for both immunology and public health. Population heterogeneities in underlying (pre-vaccination) susceptibility and vaccine response can cause measured vaccine effectiveness (mVE) to change over time even in the absence of pathogen evolution and any actual waning of immune responses. We use a multi-scale agent-based models parameterized using epidemiological and immunological data, to investigate the effect of these heterogeneities on mVE as measured by the hazard ratio. Based on our previous work, we consider waning of antibodies according to a power law and link it to protection in two ways: 1) motivated by correlates of risk data and 2) using a within-host model of stochastic viral extinction. The effect of the heterogeneities is given by concise and understandable formulas, one of which is essentially a generalization of Fisher's fundamental theorem of natural selection to include higher derivatives. Heterogeneity in underlying susceptibility accelerates apparent waning, whereas heterogeneity in vaccine response slows down apparent waning. Our models suggest that heterogeneity in underlying susceptibility is likely to dominate. However, heterogeneity in vaccine response offsets <10% to >100% (median of 29%) of this effect in our simulations. Our methodology and results may be helpful in understanding competing heterogeneities and waning of immunity and vaccine-induced protection. Our study suggests heterogeneity is more likely to 'bias' mVE downwards towards faster waning of immunity but a subtle bias in the opposite direction is also plausible.
Collapse
Affiliation(s)
- Ariel Nikas
- Emory University School of Medicine, Department of Microbiology and Immunology, Atlanta, Georgia, United States of America
| | - Hasan Ahmed
- Emory University, Department of Biology, Atlanta, Georgia, United States of America
| | - Veronika I. Zarnitsyna
- Emory University School of Medicine, Department of Microbiology and Immunology, Atlanta, Georgia, United States of America
| |
Collapse
|
38
|
Nnaji A, Ozdal MA. Perception and awareness towards malaria vaccine policy implementation in Nigeria by health policy actors. Malar J 2023; 22:111. [PMID: 36991411 PMCID: PMC10054212 DOI: 10.1186/s12936-023-04536-z] [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: 11/03/2022] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND This study aimed to assess the perception and awareness of malaria vaccine policy implementation among health policy actors in Nigeria. METHODS A descriptive study was conducted to assess the opinions and perceptions of policy actors on the implementation of a vaccination programme against malaria in Nigeria. Descriptive statistics were carried out to study the characteristics of the population and the univariate analysis of the responses to questions presented to the participants. Multinomial logistic regression was conducted to evaluate the association between demographic characteristics and the responses. RESULTS The study revealed that malaria vaccine awareness was poor, with only 48.9% of the policy actors having previous knowledge of the malaria vaccine. The majority of participants (67.8%) declared that they were aware of the importance of vaccine policy in efforts to manage disease transmission. As the number of years of work experience of the participants increased, the odds of being more likely to be aware of the malaria vaccine increased [OR 2.491 (1.183-5.250), p value < 0.05]. CONCLUSION It is recommended that policy-makers develop methods of educating populations, increase awareness of the acceptability of the vaccine and ensure that an affordable malaria vaccine programme is implemented in the population.
Collapse
Affiliation(s)
- Adaugo Nnaji
- Institute of Graduate Studies and Research, European University of Lefke, Northern Cyprus, TR-10, Mersin, Turkey.
| | - Macide Artac Ozdal
- Faculty of Health Sciences, Department of Health Management, European University of Lefke, Northern Cyprus, TR-10, Mersin, Turkey
| |
Collapse
|
39
|
Yihunie W, Kebede B, Tegegne BA, Getachew M, Abebe D, Aschale Y, Belew H, Bahiru B. Systematic Review of Safety of RTS,S with AS01 and AS02 Adjuvant Systems Using Data from Randomized Controlled Trials in Infants, Children, and Adults. Clin Pharmacol 2023; 15:21-32. [PMID: 36941908 PMCID: PMC10024506 DOI: 10.2147/cpaa.s400155] [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: 12/15/2022] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Background Emergence of antimalarial drugs and insecticides resistance alarms scientists to develop a safe and effective malaria vaccine. A pre-erythrocytic malaria vaccine called RTS,S has made great strides. Aim The review was aimed to assess the safety of the candidate malaria vaccine RTS,S with AS01 and AS02 adjuvants using data from Phase I-III randomized controlled clinical trials (RCTs). Methods This systematic review was conducted based on PRISMA 2020. Regardless of time of publication year, all articles related with safety of RTS,S, RCTs published in the English language were included in the study. The last search of databases, and registry was conducted on 30 May, 2022. Pubmed, Google Scholar, Cochrane Library, Wiley Online Library, and Clinical trials.gov were thoroughly searched for accessible RCTs on the safety of RTS,S malaria vaccine. The studies were screened in three steps: duplicate removal, title and abstract screening, and full-text review. The included studies' bias risk was assessed using the Cochrane risk of bias tool for RCTs. This systematic review is registered at Prospero (registration number: CRD42021285888). The qualitative descriptive findings from the included published studies were reported stratified by clinical trial phases. Findings A total of thirty-five eligible safety studies were identified. Injection site pain and swelling, febrile convulsion, fever, headache, meningitis, fatigue, gastroenteritis, myalgia, pneumonia, reactogenicity, and anemia were the most commonly reported adverse events. Despite few clinical trials reported serious adverse events, none of them were related to vaccination. Conclusion Most of the adverse events observed from RTS,S/AS01 and RTS,S/AS02 malaria vaccines were reported in the control group and shared by other vaccines. Hence, the authors concluded that both RTS,S/AS01 and RTS,S/AS02 malaria vaccines are safe.
Collapse
Affiliation(s)
- Wubetu Yihunie
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Bekalu Kebede
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Bantayehu Addis Tegegne
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Melese Getachew
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Dehnnet Abebe
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Yibeltal Aschale
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Habtamu Belew
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Bereket Bahiru
- Department of Pharmacy, College of Medicine and health sciences, Bahir Dar University, Bahir Dar, Ethiopia
| |
Collapse
|
40
|
Chakraborty S, Biswas S. Structure-Based Optimization of Protease-Inhibitor Interactions to Enhance Specificity of Human Stefin-A against Falcipain-2 from the Plasmodium falciparum 3D7 Strain. Biochemistry 2023; 62:1053-1069. [PMID: 36763907 DOI: 10.1021/acs.biochem.2c00585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
The emergence of resistance in Plasmodium falciparum to frontline artemisinin-based combination therapies has raised global concerns and emphasized the identification of new drug targets for malaria. Cysteine protease falcipain-2 (FP2), involved in host hemoglobin degradation and instrumental in parasite survival, has long been proposed as a promising malarial drug target. However, designing active-site-targeted small-molecule inhibitors of FP2 becomes challenging due to their off-target specificity toward highly homologous human cysteine cathepsins. The use of proteinaceous inhibitors, which have nonconserved exosite interactions and larger interface area, can effectively circumvent this problem. In this study, we report for the first time that human stefin-A (STFA) efficiently inhibits FP2 with Ki values in the nanomolar range. The FP2-STFA complex crystal structure, determined in this study, and sequence analyses identify a unique nonconserved exosite interaction, compared to human cathepsins. Designing a mutation Lys68 > Arg in STFA amplifies its selectivity garnering a 3.3-fold lower Ki value against FP2, and the crystal structure of the FP2-STFAK68R complex shows stronger electrostatic interaction between side-chains of Arg68 (STFAK68R) and Asp109 (FP2). Comparative structural analyses and molecular dynamics (MD) simulation studies of the complexes further confirm higher buried surface areas, better interaction energies for FP2-STFAK68R, and consistency of the newly developed electrostatic interaction (STFA-R68-FP2-D109) in the MD trajectory. The STFA-K68R mutant also shows higher Ki values against human cathepsin-L and stefin, a step toward eliminating off-target specificity. Hence, this work underlines the design of host-based proteinaceous inhibitors against FP2, with further optimization to render them more potent and selective.
Collapse
Affiliation(s)
- Subhoja Chakraborty
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, HBNI, 1/AF Bidhan Nagar, Kolkata 700064, India.,Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Sampa Biswas
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, HBNI, 1/AF Bidhan Nagar, Kolkata 700064, India.,Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| |
Collapse
|
41
|
Nikas A, Ahmed H, Zarnitsyna VI. Estimating Waning of Vaccine Effectiveness: A Simulation Study. Clin Infect Dis 2023; 76:479-486. [PMID: 36056892 PMCID: PMC10169445 DOI: 10.1093/cid/ciac725] [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: 04/06/2022] [Revised: 07/27/2022] [Accepted: 08/31/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Developing accurate and reliable methods to estimate vaccine protection is a key goal in immunology and public health. While several statistical methods have been proposed, their potential inaccuracy in capturing fast intraseasonal waning of vaccine-induced protection needs to be rigorously investigated. METHODS To compare statistical methods for estimating vaccine effectiveness (VE), we generated simulated data using a multiscale, agent-based model of an epidemic with an acute viral infection and differing extents of VE waning. We apply a previously proposed framework for VE measures based on the observational data richness to assess changes of vaccine-induced protection over time. RESULTS While VE measures based on hard-to-collect information (eg, the exact timing of exposures) were accurate, usually VE studies rely on time-to-infection data and the Cox proportional hazards model. We found that its extension using scaled Schoenfeld residuals, previously proposed for capturing VE waning, was unreliable in capturing both the degree of waning and its functional form and identified the mathematical factors contributing to this unreliability. We showed that partitioning time and including a time-vaccine interaction term in the Cox model significantly improved estimation of VE waning, even in the case of dramatic, rapid waning. We also proposed how to optimize the partitioning scheme. CONCLUSIONS While appropriate for rejecting the null hypothesis of no waning, scaled Schoenfeld residuals are unreliable for estimating the degree of waning. We propose a Cox-model-based method with a time-vaccine interaction term and further optimization of partitioning time. These findings may guide future analysis of VE waning data.
Collapse
Affiliation(s)
- Ariel Nikas
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Hasan Ahmed
- Department of Biology, Emory University, Atlanta, Georgia, USA
| | - Veronika I Zarnitsyna
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
42
|
Niaré K, Chege T, Rosenkranz M, Mwai K, Saßmannshausen Z, Odera D, Nyamako L, Tuju J, Alfred T, Waitumbi JN, Ogutu B, Sirima SB, Awandare G, Kouriba B, Rayner JC, Osier FHA. Characterization of a novel Plasmodium falciparum merozoite surface antigen and potential vaccine target. Front Immunol 2023; 14:1156806. [PMID: 37122725 PMCID: PMC10140549 DOI: 10.3389/fimmu.2023.1156806] [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: 02/01/2023] [Accepted: 03/31/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Detailed analyses of genetic diversity, antigenic variability, protein localization and immunological responses are vital for the prioritization of novel malaria vaccine candidates. Comprehensive approaches to determine the most appropriate antigen variants needed to provide broad protection are challenging and consequently rarely undertaken. Methods Here, we characterized PF3D7_1136200, which we named Asparagine-Rich Merozoite Antigen (ARMA) based on the analysis of its sequence, localization and immunogenicity. We analyzed IgG and IgM responses against the common variants of ARMA in independent prospective cohort studies in Burkina Faso (N = 228), Kenya (N = 252) and Mali (N = 195) using a custom microarray, Div-KILCHIP. Results We found a marked population structure between parasites from Africa and Asia. African isolates shared 34 common haplotypes, including a dominant pair although the overall selection pressure was directional (Tajima's D = -2.57; Fu and Li's F = -9.69; P < 0.02). ARMA was localized to the merozoite surface, IgG antibodies induced Fc-mediated degranulation of natural killer cells and strongly inhibited parasite growth in vitro. We found profound serological diversity, but IgG and IgM responses were highly correlated and a hierarchical clustering analysis identified only three major serogroups. Protective IgG and IgM antibodies appeared to target both cross-reactive and distinct epitopes across variants. However, combinations of IgG and IgM antibodies against selected variants were associated with complete protection against clinical episodes of malaria. Discussion Our systematic strategy exploits genomic data to deduce the handful of antigen variants with the strongest potential to induce broad protection and may be broadly applicable to other complex pathogens for which effective vaccines remain elusive.
Collapse
Affiliation(s)
- Karamoko Niaré
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
- Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Centre for Geographic Medicine Research—Coast, Kilifi, Kenya
- Malaria Research and Training Centre (MRTC), Department of Epidemiology of Parasitic Diseases, Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
- *Correspondence: Karamoko Niaré, ; Faith H. A. Osier,
| | - Timothy Chege
- Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Centre for Geographic Medicine Research—Coast, Kilifi, Kenya
| | - Micha Rosenkranz
- Centre for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Kennedy Mwai
- Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Centre for Geographic Medicine Research—Coast, Kilifi, Kenya
- Epidemiology and Biostatistics Division, School of Public Health, University of the Witwatersrand, Johannesburg, South Africa
| | - Zoe Saßmannshausen
- Centre for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Dennis Odera
- Centre for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Lydia Nyamako
- Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Centre for Geographic Medicine Research—Coast, Kilifi, Kenya
| | - James Tuju
- Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Centre for Geographic Medicine Research—Coast, Kilifi, Kenya
| | - Tiono Alfred
- Public Health Department, Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou, Burkina Faso
| | - John N. Waitumbi
- Basic Science Laboratory, US Army Medical Research Directorate-Africa/Kenya Medical Research Institute, Kisumu, Kenya
| | - Bernhards Ogutu
- Kenya Medical Research Institute, Centre for Clinical Research, Nairobi, Kenya
| | | | - Gordon Awandare
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
| | - Bourema Kouriba
- Malaria Research and Training Centre (MRTC), Department of Epidemiology of Parasitic Diseases, Faculty of Pharmacy, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
- Centre d’Infectiologie Charles Mérieux-Mali, Bamako, Mali
| | - Julian C. Rayner
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Faith H. A. Osier
- Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Centre for Geographic Medicine Research—Coast, Kilifi, Kenya
- Centre for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
- *Correspondence: Karamoko Niaré, ; Faith H. A. Osier,
| |
Collapse
|
43
|
Ngulube P. Humoral Immune Responses to P. falciparum Circumsporozoite Protein (Pfcsp) Induced by the RTS, S Vaccine - Current Update. Infect Drug Resist 2023; 16:2147-2157. [PMID: 37077252 PMCID: PMC10106824 DOI: 10.2147/idr.s401247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/23/2023] [Indexed: 04/21/2023] Open
Abstract
Malaria vaccines targeting the circumsporozoite protein (CSP) of the P. falciparum parasite have been overall relatively promising. RTS, S is a pre-erythrocytic recombinant protein-based malaria vaccine that targets CSP. RTS, S effectiveness shows some limited success regardless of its 58% efficacy for severe disease. P. falciparum circumsporozoite protein (Pfcsp) has stood to be the main candidate protein for most pre-erythrocytic stage vaccines. Studies on the structural and biophysical characteristics of antibodies specific to CSP (anti-CSP) are underway to achieve fine specificity with the CSP polymorphic regions. More recent studies have proposed the use of different kinds of monoclonal antibodies, the use of appropriate adjuvants, ideal vaccination dose and frequency, and improved targeting of particular epitopes for the robust production of functional antibodies and high complement-fixing activity as other potential methods for achieving long-lasting RTS, S. This review highlights recent findings regarding humoral immune responses to CSP elicited by RTS, S vaccine.
Collapse
Affiliation(s)
- Peter Ngulube
- Department of Biological Sciences, Academy of Medical Sciences, Malawi University of Science and Technology, Thyolo, Malawi
- Correspondence: Peter Ngulube, Email
| |
Collapse
|
44
|
Morter R, Tiono AB, Nébié I, Hague O, Ouedraogo A, Diarra A, Viebig NK, Hill AVS, Ewer KJ, Sirima SB. Impact of exposure to malaria and nutritional status on responses to the experimental malaria vaccine ChAd63 MVA ME-TRAP in 5-17 month-old children in Burkina Faso. Front Immunol 2022; 13:1058227. [PMID: 36532031 PMCID: PMC9755991 DOI: 10.3389/fimmu.2022.1058227] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
The experimental malaria vaccine ChAd63 MVA ME-TRAP previously showed protective efficacy against Plasmodium falciparum infection in Phase IIa sporozoite challenge studies in adults in the United Kingdom and in a Phase IIb field efficacy trial in Kenyan adults. However, it failed to demonstrate efficacy in a phase IIb trial in 5-17 month-old children in an area of high malaria transmission in Burkina Faso. This secondary analysis investigated whether exposure to malaria or nutritional status might be associated with reduced responses to vaccination in this cohort. Parasite blood smears and anti-AMA-1 IgG titres were used to assess history of exposure to malaria and weight-for-length Z scores were calculated to assess nutritional status. Differences in vaccine-specific anti-TRAP IgG titre and ex vivo IFNγ ELISpot response were measured between groups. In total, n = 336 volunteers randomised to receive the experimental vaccine regimen were included in this analysis. A positive smear microscopy result was associated with reduced anti-TRAP IgG titre (geometric mean titre: 2775 (uninfected) vs 1968 (infected), p = 0.025), whilst anti-AMA-1 IgG titres were weakly negatively correlated with reduced ex vivo IFNγ ELISpot response (r = -0.18, p = 0.008). Nutritional status was not associated with either humoral or cellular immunogenicity. Vaccine efficacy was also measured separately for vaccinees with positive and negative blood smears. Although not significant in either group compared to controls, vaccine efficacy measured by Cox hazard ratio was higher in uninfected compared to infected individuals (19.8% [p = 0.50] vs 3.3% [p = 0.69]). Overall, this data suggests exposure to malaria may be associated with impaired vaccine immunogenicity. This may have consequences for the testing and eventual deployment of various vaccines, in areas with high endemicity for malaria. Trial Registration Pactr.org, identifier PACTR201208000404131; ClinicalTrials.gov, identifier NCT01635647.
Collapse
Affiliation(s)
- Richard Morter
- Nuffield Department of Clinical Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Alfred B. Tiono
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso,Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Issa Nébié
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Oliver Hague
- Nuffield Department of Clinical Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Amidou Diarra
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Nicola K. Viebig
- European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Heidelberg, Germany
| | - Adrian V. S. Hill
- Nuffield Department of Clinical Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Katie J. Ewer
- Nuffield Department of Clinical Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom,*Correspondence: Sodiomon B. Sirima, ; Katie J. Ewer,
| | - Sodiomon B. Sirima
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso,*Correspondence: Sodiomon B. Sirima, ; Katie J. Ewer,
| |
Collapse
|
45
|
Friedman-Klabanoff DJ, Birkhold M, Short MT, Wilson TR, Meneses CR, Lacsina JR, Oliveira F, Kamhawi S, Valenzuela JG, Hunsberger S, Mateja A, Stoloff G, Pleguezuelos O, Memoli MJ, Laurens MB. Safety and immunogenicity of AGS-v PLUS, a mosquito saliva peptide vaccine against arboviral diseases: A randomized, double-blind, placebo-controlled Phase 1 trial. EBioMedicine 2022; 86:104375. [PMID: 36436281 PMCID: PMC9700263 DOI: 10.1016/j.ebiom.2022.104375] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Immunity to mosquito salivary proteins could provide protection against multiple mosquito-borne diseases and significantly impact public health. We evaluated the safety and immunogenicity of AGS-v PLUS, a mosquito salivary peptide vaccine, in healthy adults 18-50 years old. METHODS We conducted a randomized, double-blind, placebo-controlled Phase 1 study of AGS-v PLUS administered subcutaneously on Days 1 and 22 at the Center for Vaccine Development and Global Health, Baltimore, MD, USA. Participants were block randomized 1:1:1:1:1 to two doses saline placebo, two doses AGS-v PLUS, AGS-v PLUS/ISA-51 and saline placebo, two doses AGS-v PLUS/ISA-51, or two doses AGS-v PLUS/Alhydrogel. Primary endpoints were safety (all participants receiving ≥1 injection) and antibody and cytokine responses (all participants with day 43 samples), analysed by intention to treat. FINDINGS Between 26 August 2019 and 25 February 2020, 51 participants were enrolled and randomized, 11 into the single dose AGS-v PLUS/ISA-51 group and ten in other groups. Due to COVID-19, 15 participants did not return for day 43 samplings. Participants experienced no treatment-emergent or serious adverse events. All solicited symptoms in 2/10 placebo recipients and 22/41 AGS-v PLUS recipients after dose one and 1/10 placebo recipients and 22/41 AGS-v PLUS recipients after dose two were mild/moderate except for one severe fever the day after vaccination (placebo group). Only injection site pain was more common in vaccine groups (15/51 after dose 1 and 11/51 after dose 2) versus placebo. Compared to placebo, all vaccine groups had significantly greater fold change in anti-AGS-v PLUS IgG and IFN-ɣ from baseline. INTERPRETATION AGS-v PLUS had favourable safety profile and induced robust immune responses. Next steps will determine if findings translate into clinical efficacy against mosquito-borne diseases. FUNDING UK Department of Health and Social Care.
Collapse
Affiliation(s)
- DeAnna J Friedman-Klabanoff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Megan Birkhold
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mara T Short
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Timothy R Wilson
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Claudio R Meneses
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Joshua R Lacsina
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Fabiano Oliveira
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Shaden Kamhawi
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Jesus G Valenzuela
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Sally Hunsberger
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Allyson Mateja
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | | | - Matthew J Memoli
- Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Matthew B Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
46
|
Affiliation(s)
- Peter J Hotez
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Mogomotsi Matshaba
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
- Botswana-Baylor Children's Clinical Centre of Excellence, Gaborone, Botswana
| |
Collapse
|
47
|
Taylor SM, Korwa S, Wu A, Green CL, Freedman B, Clapp S, Kirui JK, O’Meara WP, Njuguna FM. Monthly sulfadoxine/pyrimethamine-amodiaquine or dihydroartemisinin-piperaquine as malaria chemoprevention in young Kenyan children with sickle cell anemia: A randomized controlled trial. PLoS Med 2022; 19:e1004104. [PMID: 36215323 PMCID: PMC9591057 DOI: 10.1371/journal.pmed.1004104] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 10/24/2022] [Accepted: 08/26/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Children with sickle cell anemia (SCA) in areas of Africa with endemic malaria transmission are commonly prescribed malaria chemoprevention. Chemoprevention regimens vary between countries, and the comparative efficacy of prevention regimens is largely unknown. METHODS AND FINDINGS We enrolled Kenyan children aged 1 to 10 years with homozygous hemoglobin S (HbSS) in a randomized, open-label trial conducted between January 23, 2018, and December 15, 2020, in Homa Bay, Kenya. Children were assigned 1:1:1 to daily Proguanil (the standard of care), monthly sulfadoxine/pyrimethamine-amodiaquine (SP-AQ), or monthly dihydroartemisinin-piperaquine (DP) and followed monthly for 12 months. The primary outcome was the cumulative incidence of clinical malaria at 12 months, and the main secondary outcome was the cumulative incidence of painful events by self-report. Secondary outcomes included other parasitologic, hematologic, and general events. Negative binomial models were used to estimate incidence rate ratios (IRRs) per patient-year (PPY) at risk relative to Proguanil. The primary analytic population was the As-Treated population. A total of 246 children were randomized to daily Proguanil (n = 81), monthly SP-AQ (n = 83), or monthly DP (n = 82). Overall, 53.3% (n = 131) were boys and the mean age was 4.6 ± 2.5 years. The clinical malaria incidence was 0.04 episodes/PPY; relative to the daily Proguanil group, incidence rates were not significantly different in the monthly SP-AQ (IRR: 3.05, 95% confidence interval [CI]: 0.36 to 26.14; p = 0.39) and DP (IRR: 1.36, 95% CI: 0.21 to 8.85; p = 0.90) groups. Among secondary outcomes, relative to the daily Proguanil group, the incidence of painful events was not significantly different in the monthly SP-AQ and DP groups, while monthly DP was associated with a reduced rate of dactylitis (IRR: 0.47; 95% CI: 0.23 to 0.96; p = 0.038). The incidence of Plasmodium falciparum infection relative to daily Proguanil was similar in the monthly SP-AQ group (IRR 0.46; 95% CI: 0.17 to 1.20; p = 0.13) but reduced with monthly DP (IRR 0.21; 95% CI: 0.08 to 0.56; p = 0.002). Serious adverse events were common and distributed between groups, although compared to daily Proguanil (n = 2), more children died receiving monthly SP-AQ (n = 7; hazard ratio [HR] 5.44; 95% CI: 0.92 to 32.11; p = 0.064) but not DP (n = 1; HR 0.61; 95% CI 0.04 to 9.22; p = 0.89), although differences did not reach statistical significance for either SP-AQ or DP. Study limitations include the unexpectedly limited transmission of P. falciparum in the study setting, the high use of hydroxyurea, and the enhanced supportive care for trial participants, which may limit generalizability to higher-transmission settings where routine sickle cell care is more limited. CONCLUSIONS In this study with limited malaria transmission, malaria chemoprevention in Kenyan children with SCA with monthly SP-AQ or DP did not reduce clinical malaria, but DP was associated with reduced dactylitis and P. falciparum parasitization. Pragmatic studies of chemoprevention in higher malaria transmission settings are warranted. TRIAL REGISTRATION clinicaltrials.gov (NCT03178643). Pan-African Clinical Trials Registry: PACTR201707002371165.
Collapse
Affiliation(s)
- Steve M. Taylor
- Division of Infectious Diseases, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Clinical Research Institute, Durham, North Carolina, United States of America
- Duke Global Health Institute, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| | - Sarah Korwa
- Academic Model Providing Access to Healthcare (AMPATH), Eldoret, Kenya
| | - Angie Wu
- Duke Clinical Research Institute, Durham, North Carolina, United States of America
| | - Cynthia L. Green
- Duke Clinical Research Institute, Durham, North Carolina, United States of America
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Betsy Freedman
- Division of Infectious Diseases, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Sheila Clapp
- Duke Clinical Research Institute, Durham, North Carolina, United States of America
| | | | - Wendy P. O’Meara
- Division of Infectious Diseases, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Global Health Institute, Duke University, Durham, North Carolina, United States of America
| | - Festus M. Njuguna
- Academic Model Providing Access to Healthcare (AMPATH), Eldoret, Kenya
- Department of Child Health and Paediatrics, School of Medicine, College of Health Sciences, Moi University, Eldoret, Kenya
| |
Collapse
|
48
|
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
|
49
|
Somanathan A, Mian SY, Chaddha K, Uchoi S, Bharti PK, Tandon R, Gaur D, Chauhan VS. Process development and preclinical evaluation of a major Plasmodium falciparum blood stage vaccine candidate, Cysteine-Rich Protective Antigen (CyRPA). Front Immunol 2022; 13:1005332. [PMID: 36211427 PMCID: PMC9535676 DOI: 10.3389/fimmu.2022.1005332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
Plasmodium falciparum Cysteine-Rich Protective Antigen (CyRPA) is an essential, highly conserved merozoite antigen that forms an important multi-protein complex (RH5/Ripr/CyRPA) necessary for erythrocyte invasion. CyRPA is a promising blood-stage vaccine target that has been shown to elicit potent strain-transcending parasite neutralizing antibodies. Recently, we demonstrated that naturally acquired immune anti-CyRPA antibodies are invasion-inhibitory and therefore a correlate of protection against malaria. Here, we describe a process for the large-scale production of tag-free CyRPA vaccine in E. coli and demonstrate its parasite neutralizing efficacy with commonly used adjuvants. CyRPA was purified from inclusion bodies using a one-step purification method with high purity (>90%). Biochemical and biophysical characterization showed that the purified tag-free CyRPA interacted with RH5, readily detected by a conformation-specific CyRPA monoclonal antibody and recognized by sera from malaria infected individuals thus indicating that the recombinant antigen was correctly folded and retained its native conformation. Tag-free CyRPA formulated with Freund’s adjuvant elicited highly potent parasite neutralizing antibodies achieving inhibition of >90% across diverse parasite strains. Importantly, we identified tag-free CyRPA/Alhydrogel formulation as most effective in inducing a highly immunogenic antibody response that exhibited efficacious, cross-strain in vitro parasite neutralization achieving ~80% at 10 mg/ml. Further, CyRPA/Alhydrogel vaccine induced anti-parasite cytokine response in mice. In summary, our study provides a simple, scalable, cost-effective process for the production of tag-free CyRPA that in combination with human-compatible adjuvant induces efficacious humoral and cell-mediated immune response.
Collapse
Affiliation(s)
- Anjali Somanathan
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Syed Yusuf Mian
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Kritika Chaddha
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Seemalata Uchoi
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Praveen K. Bharti
- ICMR-National Institute of Research in Tribal Health (NIRTH), Jabalpur, India
| | - Ravi Tandon
- Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Deepak Gaur
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Virander Singh Chauhan
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
- *Correspondence: Virander Singh Chauhan,
| |
Collapse
|
50
|
Owalla TJ, Hergott DEB, Seilie AM, Staubus W, Chavtur C, Chang M, Kublin JG, Egwang TG, Murphy SC. Rethinking detection of pre-existing and intervening Plasmodium infections in malaria clinical trials. Front Immunol 2022; 13:1003452. [PMID: 36203582 PMCID: PMC9531235 DOI: 10.3389/fimmu.2022.1003452] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/22/2022] [Indexed: 02/02/2023] Open
Abstract
Pre-existing and intervening low-density Plasmodium infections complicate the conduct of malaria clinical trials. These infections confound infection detection endpoints, and their immunological effects may detract from intended vaccine-induced immune responses. Historically, these infections were often unrecognized since infrequent and often analytically insensitive parasitological testing was performed before and during trials. Molecular diagnostics now permits their detection, but investigators must weigh the cost, complexity, and personnel demands on the study and the laboratory when scheduling such tests. This paper discusses the effect of pre-existing and intervening, low-density Plasmodium infections on malaria vaccine trial endpoints and the current methods employed for their infection detection. We review detection techniques, that until recently, provided a dearth of cost-effective strategies for detecting low density infections. A recently deployed, field-tested, simple, and cost-effective molecular diagnostic strategy for detecting pre-existing and intervening Plasmodium infections from dried blood spots (DBS) in malaria-endemic settings is discussed to inform new clinical trial designs. Strategies that combine sensitive molecular diagnostic techniques with convenient DBS collections and cost-effective pooling strategies may enable more thorough and informative infection monitoring in upcoming malaria clinical trials and epidemiological studies.
Collapse
Affiliation(s)
- Tonny J. Owalla
- Department of Immunology and Parasitology, Med Biotech Laboratories, Kampala, Uganda
| | - Dianna E. B. Hergott
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, United States
| | - Annette M. Seilie
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Weston Staubus
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Chris Chavtur
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Ming Chang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - James G. Kublin
- Department of Global Health, University of Washington, Seattle, WA, United States,Seattle Malaria Clinical Trials Center, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Thomas G. Egwang
- Department of Immunology and Parasitology, Med Biotech Laboratories, Kampala, Uganda
| | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States,Seattle Malaria Clinical Trials Center, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States,Department of Microbiology, University of Washington, Seattle, WA, United States,*Correspondence: Sean C. Murphy,
| |
Collapse
|