1
|
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
|
2
|
Ge Q, Chen P, Cheng Y, Xiao Y. The long road for vaccine development with difficulties and hopes. Emerg Microbes Infect 2024; 13:2396886. [PMID: 39178195 PMCID: PMC11378663 DOI: 10.1080/22221751.2024.2396886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 08/25/2024]
Affiliation(s)
- Qi Ge
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Pan Chen
- Peking Union Medical College & Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Yan Cheng
- Peking Union Medical College & Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Yonghong Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Peking Union Medical College & Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Department of Structure and Morphology, Jinan Microecological Biomedicine Shandong Laboratory, Jinan, People's Republic of China
| |
Collapse
|
3
|
Andrews KT, Fisher GM, Firmin M, Liepa AJ, Wilson T, Gardiner J, Mohri Y, Debele E, Rai A, Davey AK, Masurier A, Delion A, Mouratidis AA, Hutt OE, Forsyth CM, Burrows JN, Ryan JH, Riches AG, Skinner-Adams TS. Discovery of 1,3,4-oxadiazoles with slow-action activity against Plasmodium falciparum malaria parasites. Eur J Med Chem 2024; 278:116796. [PMID: 39241483 DOI: 10.1016/j.ejmech.2024.116796] [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: 04/05/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/09/2024]
Abstract
To achieve malaria eradication, new preventative agents that act differently to front-line treatment drugs are needed. To identify potential chemoprevention starting points we screened a sub-set of the CSIRO Australia Compound Collection for compounds with slow-action in vitro activity against Plasmodium falciparum. This work identified N,N-dialkyl-5-alkylsulfonyl-1,3,4-oxadiazol-2-amines as a new antiplasmodial chemotype (e.g., 1 96 h IC50 550 nM; 3 96 h IC50 160 nM) with a different action to delayed-death slow-action drugs. A series of analogues were synthesized from thiotetrazoles and carbomoyl derivatives using Huisgen 1,3,4-oxadiazole synthesis followed by oxidation of the resultant thioethers to target sulfones. Structure activity relationship analysis of analogues identified compounds with potent and selective in vitro activity against drug-sensitive and multi-drug resistant Plasmodium parasites (e.g., 31 and 32 96 h IC50 <40 nM; SI > 2500). Subsequent studies in mice with compound 1, which had the best microsomal stability of the compounds assessed (T1/2 >255 min), demonstrated rapid clearance and poor oral in vivo efficacy in a P. berghei murine malaria model. These data indicate that while N,N-dialkyl-5-alkylsulfonyl-1,3,4-oxadiazol-2-amines are a novel class of slow-acting antiplasmodial agents, the further development of this chemotype for malaria chemoprophylaxis will require pharmacokinetic profile improvements.
Collapse
Affiliation(s)
- Katherine T Andrews
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, Queensland, 4111, Australia.
| | - Gillian M Fisher
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, Queensland, 4111, Australia
| | - Meaghan Firmin
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, Queensland, 4111, Australia
| | - Andris J Liepa
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, Victoria, 3168, Australia
| | - Tony Wilson
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, Victoria, 3168, Australia
| | - James Gardiner
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, Victoria, 3168, Australia
| | - Yacine Mohri
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, Victoria, 3168, Australia
| | - Emmanuel Debele
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, Victoria, 3168, Australia
| | - Anjana Rai
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, Queensland, 4111, Australia
| | - Andrew K Davey
- Griffith Health Group, Griffith University, Gold Coast, Queensland, 4222, Australia
| | - Antoine Masurier
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, Victoria, 3168, Australia
| | - Alix Delion
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, Victoria, 3168, Australia
| | - Alexandros A Mouratidis
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, Victoria, 3168, Australia
| | - Oliver E Hutt
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, Victoria, 3168, Australia
| | - Craig M Forsyth
- School of Chemistry, Monash University, Clayton, Victoria, 3800, Australia
| | | | - John H Ryan
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, Victoria, 3168, Australia.
| | - Andrew G Riches
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, Victoria, 3168, Australia.
| | - Tina S Skinner-Adams
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, Queensland, 4111, Australia.
| |
Collapse
|
4
|
Mitchell RA, Macià D, Jairoce C, Mpina M, Naidu A, Chopo-Pizarro A, Vázquez-Santiago M, Campo JJ, Aide P, Aguilar R, Daubenberger C, Dobaño C, Moncunill G. Effect of RTS,S/AS01 E vaccine booster dose on cellular immune responses in African infants and children. NPJ Vaccines 2024; 9:200. [PMID: 39455625 PMCID: PMC11511852 DOI: 10.1038/s41541-024-00977-y] [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: 05/08/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
RTS,S/AS01E, the first approved malaria vaccine, demonstrated moderate efficacy during the phase 3 pediatric trial. We previously investigated cell-mediated immune (CMI) responses following the primary 3-dose immunization and now report responses to the booster dose given 18 months later. Thirty CMI markers were measured by Luminex in supernatants of peripheral blood mononuclear cells from 709 children and infants after RTS,S/AS01E antigen stimulation, and their associations with malaria risk and antibodies one month post-booster and one year later were assessed. IL-2, IFN-γ, IL-17, IL-5, and IL-13 were associated with RTS,S/AS01E booster vaccination, and IL-2 responses to the circumsporozoite protein (CSP) remained higher after one year. IL-2 was associated with reduced malaria risk in one site, and IL-10 was associated with increased risk in infants. Anti-CSP IgG and IL-2 were moderately correlated one year after booster. This study highlights the moderate cell-mediated immunogenicity of the RTS,S/AS01E booster dose that aligns with partial recovery of RTS,S/AS01E vaccine efficacy.
Collapse
Affiliation(s)
- Robert A Mitchell
- ISGlobal, Barcelona, Catalonia, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Dídac Macià
- ISGlobal, Barcelona, Catalonia, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Chenjerai Jairoce
- ISGlobal, Barcelona, Catalonia, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Maxmillian Mpina
- Ifakara Health Institute. Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | | | | | | | | - Pedro Aide
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Ruth Aguilar
- ISGlobal, Barcelona, Catalonia, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Claudia Daubenberger
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Carlota Dobaño
- ISGlobal, Barcelona, Catalonia, Spain.
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain.
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain.
| | - Gemma Moncunill
- ISGlobal, Barcelona, Catalonia, Spain.
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain.
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain.
| |
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
|
Macià D, Campo JJ, Jairoce C, Mpina M, Sorgho H, Dosoo D, Agnandji ST, Kusi KA, Molinos-Albert LM, Kariuki S, Daubenberger C, Mordmüller B, Moncunill G, Dobaño C. The effect of Plasmodium falciparum exposure and maternal anti-circumsporozoite protein antibodies on responses to RTS,S/AS01 E vaccination in infants and children: an ancillary observational immunological study to a phase 3, randomised clinical trial. THE LANCET. INFECTIOUS DISEASES 2024:S1473-3099(24)00527-9. [PMID: 39461358 DOI: 10.1016/s1473-3099(24)00527-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND The RTS,S/AS01E malaria vaccine showed lower antibody response and protective efficacy in infants aged 6-12 weeks compared with children aged 5-17 months (for whom this vaccine is recommended). We aimed to study the effect of previous Plasmodium falciparum exposure on the antibody responses to RTS,S/AS01E vaccination in infants and children, and the mediating effect of baseline (including maternal) anti-circumsporozoite protein (CSP) antibodies. METHODS In this observational study, we included children and infants from six African countries (Burkina Faso, Gabon, Ghana, Kenya, Mozambique, and Tanzania) enrolled in the MAL067 immunology ancillary study of the RTS,S/AS01E phase 3 clinical trial from March 27, 2009, to Jan 21, 2011. We used comparator-vaccinated infants and children to identify antibody-based signatures of previous P falciparum exposure, which were later applied to RTS,S/AS01E-vaccinated infants and children. In these participants, we explored the relationship between vaccine antibody immunoglobulin G (IgG) responses measured by ELISA and pre-vaccination serological markers of malaria exposure by assessing the IgG levels against 1000 P falciparum antigens using partial proteome microarrays. FINDINGS We included 718 comparator-vaccinated infants (348 [48%]) and children (370 [52%]) and 606 RTS,S/AS01E-vaccinated infants (329 [54%]) and children (277 [46%]). Anti-CSP IgG responses to primary vaccination did not correlate with a baseline signature of previous exposure in children, suggesting that prior P falciparum exposure does not significantly affect antibody immunogenicity in children (Pearson's r=-0·02 [95% CI -0·13 to 0·10]). By contrast, high P falciparum exposure signature levels at the time of vaccination in infants, presumably driven by maternally transferred antibodies and declining within the initial 6-12 months of life, correlated with reduced RTS,S/AS01E responses (r=-0·17 [-0·27 to -0·06]). This negative correlation was stronger for anti-CSP IgG than for the exposure signature or any other more immunogenic blood stage P falciparum antigens (r=-0·42 [-0·50 to -0·33]), persisted after adjustment by baseline levels of the exposure signature (semi-partial correlation r=-0·44 [-0·55 to -0·33]), and involved antibodies to the central NANP region (r=-0·39 [-0·49 to -0·28]) but not the C-terminal region (r=0·02 [-0·10 to 0·15]) of CSP. The negative effect of maternal anti-CSP IgG in infants did not appear to be confounded by other malaria transmission-dependent variables. INTERPRETATION Interference between passive immunity and vaccine response is clinically significant and might affect the implementation of next-generation CSP-based vaccines for young infants and mothers as well as passive immunisation with human monoclonal antibodies. FUNDING US National Institutes of Health, National Institute of Allergy and Infectious Diseases; PATH-Malaria Vaccine Initiative; Spanish Ministerio de Economía y Competitividad (Instituto de Salud Carlos III), European Regional Development Fund and European Social Fund; Fundación Ramón Areces; Spanish Ministry of Science and Innovation; and Generalitat de Catalunya (CERCA Program).
Collapse
Affiliation(s)
- Dídac Macià
- ISGlobal, Barcelona, Catalonia, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | | | - Chenjerai Jairoce
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Cambeve, Vila de Manhiça, Maputo, Mozambique
| | - Maximilian Mpina
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Hermann Sorgho
- Unité de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de la Santé, Nanoro, Burkina Faso
| | - David Dosoo
- Kintampo Health Research Centre, Kintampo, Brong-Ahafo, Ghana
| | - Selidji Todagbe Agnandji
- Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon; Institute of Tropical Medicine and German Center for Infection Research, University of Tübingen, Tübingen, Germany
| | - Kwadwo Asamoah Kusi
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Luis M Molinos-Albert
- ISGlobal, Barcelona, Catalonia, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Simon Kariuki
- Kenya Medical Research Institute/Centre for Global Health, Kisumu, Kisumu, Kenya
| | - Claudia Daubenberger
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Benjamin Mordmüller
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain; Institute of Tropical Medicine and German Center for Infection Research, University of Tübingen, Tübingen, Germany; Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Gemma Moncunill
- ISGlobal, Barcelona, Catalonia, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Carlota Dobaño
- ISGlobal, Barcelona, Catalonia, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain.
| |
Collapse
|
7
|
Palacpac NMQ, Horii T. Understanding the immunogenicity of RTS,S in infants. THE LANCET. INFECTIOUS DISEASES 2024:S1473-3099(24)00590-5. [PMID: 39461357 DOI: 10.1016/s1473-3099(24)00590-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/29/2024]
Affiliation(s)
- Nirianne Marie Q Palacpac
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Toshihiro Horii
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
8
|
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
|
9
|
Spreng RL, Seaton KE, Lin L, Hilliard S, Horn GQ, Abraha M, Deal AW, Li K, Carnacchi AJ, Feeney E, Shabbir S, Zhang L, Bekker V, Mudrak SV, Dutta S, Mercer LD, Gregory S, King CR, Wille-Reece U, Jongert E, Kisalu NK, Tomaras GD, Dennison SM. Identification of RTS,S/AS01 vaccine-induced humoral biomarkers predictive of protection against controlled human malaria infection. JCI Insight 2024; 9:e178801. [PMID: 39377226 PMCID: PMC11466194 DOI: 10.1172/jci.insight.178801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 08/23/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUNDThe mechanism(s) responsible for the efficacy of WHO-recommended malaria vaccine RTS,S/AS01 are not completely understood. We previously identified RTS,S vaccine-induced Plasmodium falciparum circumsporozoite protein-specific (PfCSP-specific) antibody measures associated with protection from controlled human malaria infection (CHMI). Here, we tested the protection-predicting capability of these measures in independent CHMI studies.METHODSVaccine-induced total serum antibody (immunoglobulins, Igs) and subclass antibody (IgG1 and IgG3) responses were measured by biolayer interferometry and the binding antibody multiplex assay, respectively. Immune responses were compared between protected and nonprotected vaccinees using univariate and multivariate logistic regression.RESULTSBlinded prediction analysis showed that 5 antibody binding measures, including magnitude-avidity composite of serum Ig specific for PfCSP, major NANP repeats and N-terminal junction, and PfCSP- and NANP-specific IgG1 subclass magnitude, had good prediction accuracy (area under the receiver operating characteristic curves [ROC AUC] > 0.7) in at least 1 trial. Furthermore, univariate analysis showed a significant association between these antibody measures and protection (odds ratios 2.6-3.1). Multivariate modeling of combined data from 3 RTS,S CHMI trials identified the combination of IgG1 NANP binding magnitude plus serum NANP and N-junction Ig binding magnitude-avidity composite as the best predictor of protection (95% confidence interval for ROC AUC 0.693-0.834).CONCLUSIONThese results reinforce our previous findings and provide a tool for predicting protection in future trials.TRIAL REGISTRATIONClinicalTrials.gov NCT03162614, NCT03824236, NCT01366534, and NCT01857869.FUNDINGThis study was supported by Bill & Melinda Gates Foundation's Global Health-Discovery Collaboratory grants (INV-008612 and INV-043419) to GDT.
Collapse
Affiliation(s)
| | - Kelly E. Seaton
- Center for Human Systems Immunology
- Department of Surgery, and
| | - Lin Lin
- Center for Human Systems Immunology
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | | | - Gillian Q. Horn
- Center for Human Systems Immunology
- Department of Surgery, and
| | - Milite Abraha
- Center for Human Systems Immunology
- Department of Surgery, and
| | - Aaron W. Deal
- Center for Human Systems Immunology
- Department of Surgery, and
| | - Kan Li
- Center for Human Systems Immunology
- Department of Surgery, and
| | | | | | - Siam Shabbir
- Center for Human Systems Immunology
- Department of Surgery, and
| | - Lu Zhang
- Center for Human Systems Immunology
- Department of Surgery, and
| | - Valerie Bekker
- Center for Human Systems Immunology
- Department of Surgery, and
| | - Sarah V. Mudrak
- Center for Human Systems Immunology
- Department of Surgery, and
| | - Sheetij Dutta
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Laina D. Mercer
- Center for Vaccine Innovation and Access, PATH, Seattle, Washington, USA
| | - Scott Gregory
- Center for Vaccine Innovation and Access, PATH, Washington, DC, USA
| | - C. Richter King
- Center for Vaccine Innovation and Access, PATH, Washington, DC, USA
| | | | | | | | - Georgia D. Tomaras
- Duke Human Vaccine Institute
- Center for Human Systems Immunology
- Department of Surgery, and
- Department of Integrative Immunobiology and
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | | |
Collapse
|
10
|
Saha D, Pramanik A, Freville A, Siddiqui AA, Pal U, Banerjee C, Nag S, Debsharma S, Pramanik S, Mazumder S, Maiti NC, Datta S, van Ooij C, Bandyopadhyay U. Structure-function analysis of nucleotide housekeeping protein HAM1 from human malaria parasite Plasmodium falciparum. FEBS J 2024; 291:4349-4371. [PMID: 39003571 DOI: 10.1111/febs.17216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/29/2024] [Accepted: 06/20/2024] [Indexed: 07/15/2024]
Abstract
Non-canonical nucleotides, generated as oxidative metabolic by-products, significantly threaten the genome integrity of Plasmodium falciparum and thereby, their survival, owing to their mutagenic effects. PfHAM1, an evolutionarily conserved inosine/xanthosine triphosphate pyrophosphohydrolase, maintains nucleotide homeostasis in the malaria parasite by removing non-canonical nucleotides, although structure-function intricacies are hitherto poorly reported. Here, we report the X-ray crystal structure of PfHAM1, which revealed a homodimeric structure, additionally validated by size-exclusion chromatography-multi-angle light scattering analysis. The two monomeric units in the dimer were aligned in a parallel fashion, and critical residues associated with substrate and metal binding were identified, wherein a notable structural difference was observed in the β-sheet main frame compared to human inosine triphosphate pyrophosphatase. PfHAM1 exhibited Mg++-dependent pyrophosphohydrolase activity and the highest binding affinity to dITP compared to other non-canonical nucleotides as measured by isothermal titration calorimetry. Modifying the pfham1 genomic locus followed by live-cell imaging of expressed mNeonGreen-tagged PfHAM1 demonstrated its ubiquitous presence in the cytoplasm across erythrocytic stages with greater expression in trophozoites and schizonts. Interestingly, CRISPR-Cas9/DiCre recombinase-guided pfham1-null P. falciparum survived in culture under standard growth conditions, indicating its assistive role in non-canonical nucleotide clearance during intra-erythrocytic stages. This is the first comprehensive structural and functional report of PfHAM1, an atypical nucleotide-cleansing enzyme in P. falciparum.
Collapse
Affiliation(s)
- Debanjan Saha
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Atanu Pramanik
- Division of Structural Biology & Bioinformatics, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Aline Freville
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, UK
| | - Asim Azhar Siddiqui
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Uttam Pal
- Division of Structural Biology & Bioinformatics, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Chinmoy Banerjee
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Shiladitya Nag
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Subhashis Debsharma
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Saikat Pramanik
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Somnath Mazumder
- Department of Zoology, Raja Peary Mohan College, Uttarpara, India
| | - Nakul C Maiti
- Division of Structural Biology & Bioinformatics, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Saumen Datta
- Division of Structural Biology & Bioinformatics, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Christiaan van Ooij
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, UK
| | - Uday Bandyopadhyay
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Department of Biological Sciences, Bose Institute, Kolkata, India
| |
Collapse
|
11
|
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
|
12
|
Ali Albsheer MM, Hubbard A, Dieng CC, Gebremeskel EI, Ahmed S, Rougeron V, Ibrahim ME, Lo E, Abdel Hamid MM. Extensive genetic diversity in Plasmodium vivax from Sudan and its genetic relationships with other geographical isolates. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 123:105643. [PMID: 39053565 DOI: 10.1016/j.meegid.2024.105643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/03/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024]
Abstract
Plasmodium vivax, traditionally overlooked has experienced a notable increase in cases in East Africa. This study investigated the geographical origin and genetic diversity of P. vivax in Sudan using 14 microsatellite markers. A total of 113 clinical P. vivax samples were collected from two different ecogeographical zones, New Halfa and Khartoum, in Sudan. Additionally, 841 geographical samples from the database were incorporated for a global genetic analysis to discern genetic relationships among P. vivax isolates on regional and worldwide scales. On the regional scale, our findings revealed 91 unique and 8 shared haplotypes among the Sudan samples, showcasing a remarkable genetic diversity compared to other geographical isolates and supporting the hypothesis that P. vivax originated from Africa. On a global scale, distinct genetic clustering of P. vivax isolates from Africa, South America, and Asia (including Papua New Guinea and Solomon Island) was observed, with limited admixture among the three clusters. Principal component analysis emphasized the substantial contribution of African isolates to the observed global genetic variation. The Sudanese populations displayed extensive genetic diversity, marked by significant multi-locus linkage disequilibrium, suggesting an ancestral source of P. vivax variation globally and frequent recombination among the isolates. Notably, the East African P. vivax exhibited similarity with some Asian isolates, indicating potential recent introductions. Overall, our results underscore the effectiveness of utilizing microsatellite markers for implementing robust control measures, given their ability to capture extensive genetic diversity and linkage disequilibrium patterns.
Collapse
Affiliation(s)
- Musab M Ali Albsheer
- Department of Parasitology and Medical Entomology, Institute of Endemic Diseases, University of Khartoum, Khartoum, Sudan; Faculty of Medical Laboratory Sciences, Sinnar University, Sudan
| | - Alfred Hubbard
- Department of Bioinformatics and Genomics, University of North Carolina, Charlotte, NC 28223, USA
| | - Cheikh Cambel Dieng
- Department of Microbiology and Immunology, Drexel University, Philadelphia, PA 19129, USA
| | | | - Safaa Ahmed
- Department of Parasitology and Medical Entomology, Institute of Endemic Diseases, University of Khartoum, Khartoum, Sudan
| | - Virginie Rougeron
- Laboratoire MIVEGEC (Université de Montpellier-CNRS-IRD), CREES, 34394 Montpellier, France
| | - Muntaser E Ibrahim
- Department of Parasitology and Medical Entomology, Institute of Endemic Diseases, University of Khartoum, Khartoum, Sudan
| | - Eugenia Lo
- Department of Microbiology and Immunology, Drexel University, Philadelphia, PA 19129, USA.
| | - Muzamil M Abdel Hamid
- Department of Parasitology and Medical Entomology, Institute of Endemic Diseases, University of Khartoum, Khartoum, Sudan.
| |
Collapse
|
13
|
Sattler JM, Keiber L, Abdelrahim A, Zheng X, Jäcklin M, Zechel L, Moreau CA, Steinbrück S, Fischer M, Janse CJ, Hoffmann A, Hentzschel F, Frischknecht F. Experimental vaccination by single dose sporozoite injection of blood-stage attenuated malaria parasites. EMBO Mol Med 2024; 16:2060-2079. [PMID: 39103697 PMCID: PMC11392930 DOI: 10.1038/s44321-024-00101-6] [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: 04/27/2023] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Malaria vaccination approaches using live Plasmodium parasites are currently explored, with either attenuated mosquito-derived sporozoites or attenuated blood-stage parasites. Both approaches would profit from the availability of attenuated and avirulent parasites with a reduced blood-stage multiplication rate. Here we screened gene-deletion mutants of the rodent parasite P. berghei and the human parasite P. falciparum for slow growth. Furthermore, we tested the P. berghei mutants for avirulence and resolving blood-stage infections, while preserving sporozoite formation and liver infection. Targeting 51 genes yielded 18 P. berghei gene-deletion mutants with several mutants causing mild infections. Infections with the two most attenuated mutants either by blood stages or by sporozoites were cleared by the immune response. Immunization of mice led to protection from disease after challenge with wild-type sporozoites. Two of six generated P. falciparum gene-deletion mutants showed a slow growth rate. Slow-growing, avirulent P. falciparum mutants will constitute valuable tools to inform on the induction of immune responses and will aid in developing new as well as safeguarding existing attenuated parasite vaccines.
Collapse
Affiliation(s)
- Julia M Sattler
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, 69120, Heidelberg, Germany
- German Center for Infection Research, DZIF, Partner Site Heidelberg, Heidelberg, Germany
| | - Lukas Keiber
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, 69120, Heidelberg, Germany
- German Center for Infection Research, DZIF, Partner Site Heidelberg, Heidelberg, Germany
| | - Aiman Abdelrahim
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, 69120, Heidelberg, Germany
- German Center for Infection Research, DZIF, Partner Site Heidelberg, Heidelberg, Germany
| | - Xinyu Zheng
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, 69120, Heidelberg, Germany
- German Center for Infection Research, DZIF, Partner Site Heidelberg, Heidelberg, Germany
| | - Martin Jäcklin
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, 69120, Heidelberg, Germany
- German Center for Infection Research, DZIF, Partner Site Heidelberg, Heidelberg, Germany
| | - Luisa Zechel
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, 69120, Heidelberg, Germany
- German Center for Infection Research, DZIF, Partner Site Heidelberg, Heidelberg, Germany
| | - Catherine A Moreau
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, 69120, Heidelberg, Germany
| | - Smilla Steinbrück
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, 69120, Heidelberg, Germany
- German Center for Infection Research, DZIF, Partner Site Heidelberg, Heidelberg, Germany
| | - Manuel Fischer
- Department of Neuroradiology, Heidelberg University Medical School, 69120, Heidelberg, Germany
| | - Chris J Janse
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Angelika Hoffmann
- Department of Neuroradiology, Heidelberg University Medical School, 69120, Heidelberg, Germany
- Department of Neuroradiology, University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, 3010, Bern, Switzerland
| | - Franziska Hentzschel
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, 69120, Heidelberg, Germany
- German Center for Infection Research, DZIF, Partner Site Heidelberg, Heidelberg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, 69120, Heidelberg, Germany.
- German Center for Infection Research, DZIF, Partner Site Heidelberg, Heidelberg, Germany.
| |
Collapse
|
14
|
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
|
15
|
Ramezani-Rad P, Marina-Zárate E, Maiorino L, Myers A, Michaels KK, Pires IS, Bloom NI, Lopez PG, Cottrell CA, Burton I, Groschel B, Pradhan A, Stiegler G, Budai M, Kumar D, Pallerla S, Sayeed E, Sagar SL, Kasturi SP, Van Rompay KKA, Hangartner L, Wagner A, Burton DR, Schief WR, Crotty S, Irvine DJ. Dose-dependent regulation of immune memory responses against HIV by saponin monophosphoryl lipid A nanoparticle adjuvant. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.604373. [PMID: 39211109 PMCID: PMC11361155 DOI: 10.1101/2024.07.31.604373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The induction of durable protective immune responses is the main goal of prophylactic vaccines, and adjuvants play an important role as drivers of such responses. Despite advances in vaccine strategies, a safe and effective HIV vaccine remains a significant challenge. The use of an appropriate adjuvant is crucial to the success of HIV vaccines. Here we assessed the saponin/MPLA nanoparticle (SMNP) adjuvant with an HIV envelope (Env) trimer, evaluating the safety and impact of multiple variables including adjuvant dose (16-fold dose range), immunization route, and adjuvant composition on the establishment of Env-specific memory T and B cell responses (T Mem and B Mem ) and long-lived plasma cells in non-human primates. Robust B Mem were detected in all groups, but a 6-fold increase was observed in the highest SMNP dose group vs. the lowest dose group. Similarly, stronger vaccine responses were induced in the highest SMNP dose for CD40L + OX40 + CD4 T Mem (11-fold), IFNγ + CD4 T Mem (15-fold), IL21 + CD4 T Mem (9-fold), circulating T FH (3.6-fold), bone marrow plasma cells (7-fold), and binding IgG (1.3-fold). Substantial tier-2 neutralizing antibodies were only observed in the higher SMNP dose groups. These investigations highlight the dose-dependent potency of SMNP in non-human primates, which are relevant for human use and next-generation vaccines.
Collapse
|
16
|
Garrett N, Dintwe O, Monaco CL, Jones M, Seaton KE, Church EC, Grunenberg N, Hutter J, deCamp A, Huang Y, Lu H, Mann P, Robinson ST, Heptinstall J, Jensen RL, Pantaleo G, Ding S, Koutsoukos M, Hosseinipour MC, Van Der Meeren O, Gilbert PB, Ferrari G, Andersen-Nissen E, McElrath MJ, Tomaras GD, Gray GE, Corey L, Kublin JG. Safety and Immunogenicity of a DNA Vaccine With Subtype C gp120 Protein Adjuvanted With MF59 or AS01B: A Phase 1/2a HIV-1 Vaccine Trial. J Acquir Immune Defic Syndr 2024; 96:350-360. [PMID: 38916429 PMCID: PMC11195930 DOI: 10.1097/qai.0000000000003438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 04/02/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND An effective vaccine is required to end the HIV pandemic. We evaluated the safety and immunogenicity of a DNA (DNA-HIV-PT123) vaccine with low- or high-dose bivalent (TV1.C and 1086.C glycoprotein 120) subtype C envelope protein combinations, adjuvanted with MF59 or AS01B. METHODS HIV Vaccine Trials Network (HVTN)108 was a randomized, placebo-controlled, double-blind, phase 1/2a trial conducted in the United States and South Africa. HIV-negative adults were randomly assigned to 1 of 7 intervention arms or placebo to assess DNA prime with DNA/protein/adjuvant boosts, DNA/protein/adjuvant co-administration, and low-dose protein/adjuvant regimens. HVTN111 trial participants who received an identical regimen were also included. Outcomes included safety and immunogenicity 2 weeks and 6 months after final vaccination. RESULTS From June 2016 to July 2018, 400 participants were enrolled (N = 334 HVTN108, N = 66 HVTN111); 370 received vaccine and 30 received placebo. There were 48 grade 3 and 3 grade 4 reactogenicity events among 39/400 (9.8%) participants, and 32 mild/moderate-related adverse events in 23/400 (5.8%) participants. All intervention groups demonstrated high IgG response rates (>89%) and high magnitudes to HIV-1 Env gp120 and gp140 proteins; response rates for AS01B-adjuvanted groups approached 100%. V1V2 IgG magnitude, Fc-mediated functions, IgG3 Env response rates, and CD4+ T-cell response magnitudes and rates were higher in the AS01B-adjuvanted groups. The AS01B-adjuvanted low-dose protein elicited greater IgG responses than the higher protein dose. CONCLUSIONS The vaccine regimens were generally well tolerated. Co-administration of DNA with AS01B-adjuvanted bivalent Env gp120 elicited the strongest humoral responses; AS01B-adjuvanted regimens elicited stronger CD4+ T-cell responses, justifying further evaluation.ClinicalTrials.gov registration: NCT02915016, registered 26 September 2016.
Collapse
Affiliation(s)
- Nigel Garrett
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
- Department of Public Health Medicine, School of Nursing and Public Health, University of KwaZulu-Natal, Durban, South Africa
| | - One Dintwe
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
- Cape Town HVTN Immunology Laboratory, Cape Town, South Africa
| | - Cynthia L. Monaco
- Department of Medicine, Division of Infectious Diseases, University of Rochester Medical Center, Rochester, NY
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY
| | - Megan Jones
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Kelly E. Seaton
- Center for Human Systems Immunology, Departments of Surgery, Molecular Genetics and Microbiology, and Immunology, Duke University School of Medicine, Durham, NC
| | - E. Chandler Church
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Nicole Grunenberg
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Julia Hutter
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Allan deCamp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Huiyin Lu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Philipp Mann
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Samuel T. Robinson
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Jack Heptinstall
- Center for Human Systems Immunology, Departments of Surgery, Molecular Genetics and Microbiology, and Immunology, Duke University School of Medicine, Durham, NC
| | - Ryan L. Jensen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Giuseppe Pantaleo
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Song Ding
- EuroVacc Foundation, Lausanne, Switzerland
| | | | - Mina C. Hosseinipour
- University of North Carolina at Chapel Hill, Chapel Hill, NC
- UNC Project-Malawi, Lilongwe, Malawi
| | | | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Guido Ferrari
- Center for Human Systems Immunology, Departments of Surgery, Molecular Genetics and Microbiology, and Immunology, Duke University School of Medicine, Durham, NC
| | - Erica Andersen-Nissen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
- Cape Town HVTN Immunology Laboratory, Cape Town, South Africa
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Georgia D. Tomaras
- Center for Human Systems Immunology, Departments of Surgery, Molecular Genetics and Microbiology, and Immunology, Duke University School of Medicine, Durham, NC
| | - Glenda E. Gray
- South African Medical Research Council, Tygerberg, South Africa
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - James G. Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| |
Collapse
|
17
|
Zhu Y, Zhou L, Mo L, Hong C, Pan L, Lin J, Qi Y, Tan S, Qian M, Hu T, Zhao Y, Qiu H, Lin P, Ma X, Yang Q. Plasmodium yoelii Infection Enhances the Expansion of Myeloid-Derived Suppressor Cells via JAK/STAT3 Pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:170-186. [PMID: 38819229 DOI: 10.4049/jimmunol.2300541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 05/07/2024] [Indexed: 06/01/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs), the negative immune regulators, have been demonstrated to be involved in immune responses to a variety of pathological conditions, such as tumors, chronic inflammation, and infectious diseases. However, the roles and mechanisms underlying the expansion of MDSCs in malaria remain unclear. In this study, the phenotypic and functional characteristics of splenic MDSCs during Plasmodium yoelii NSM infection are described. Furthermore, we provide compelling evidence that the sera from P. yoelii-infected C57BL/6 mice containing excess IL-6 and granulocyte-macrophage colony-stimulating factor promote the accumulation of MDSCs by inducing Bcl2 expression. Serum-induced MDSCs exert more potent suppressive effects on T cell responses than control MDSCs within both in vivo P. yoelii infection and in vitro serum-treated bone marrow cells experiments. Serum treatment increases the MDSC inhibitory effect, which is dependent on Arg1 expression. Moreover, mechanistic studies reveal that the serum effects are mediated by JAK/STAT3 signaling. By inhibiting STAT3 phosphorylation with the JAK inhibitor JSI-124, effects of serum on MDSCs are almost eliminated. In vivo depletion of MDSCs with anti-Gr-1 or 5-fluorouracil significantly reduces the parasitemia and promotes Th1 immune response in P. yoelii-infected C57BL/6 mice by upregulating IFN-γ expression. In summary, this study indicates that P. yoelii infection facilitates the accumulation and function of MDSCs by upregulating the expression of Bcl2 and Arg1 via JAK/STAT3 signaling pathway in vivo and in vitro. Manipulating the JAK/STAT3 signaling pathway or depleting MDSCs could be promising therapeutic interventions to treat malaria.
Collapse
Affiliation(s)
- Yiqiang Zhu
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Lu Zhou
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Lengshan Mo
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Cansheng Hong
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Lingxia Pan
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jie Lin
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yanwei Qi
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Simin Tan
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Manhongtian Qian
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Tengfei Hu
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yi Zhao
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Huaina Qiu
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Peibin Lin
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Xiancai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Quan Yang
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
18
|
Sifuna PM, Mbinji M, Lucas TO, Onyango I, Akala HM, Waitumbi JN, Ogutu BR, Hutter JN, Otieno W. The Walter Reed Project, Kisumu Field Station: Impact of Research on Malaria Policy, Management, and Prevention. Am J Trop Med Hyg 2024; 110:1069-1079. [PMID: 38653233 PMCID: PMC11154051 DOI: 10.4269/ajtmh.23-0115] [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: 02/21/2023] [Accepted: 02/16/2024] [Indexed: 04/25/2024] Open
Abstract
The Walter Reed Project is a collaboration between the Walter Reed Army Institute of Research of the United States Department of Defense and the Kenya Medical Research Institute. The Kisumu field station, comprising four campuses, has until recently been devoted primarily to research on malaria countermeasures. The Kombewa Clinical Research Center is dedicated to conducting regulated clinical trials of therapeutic and vaccine candidates in development. The center's robust population-based surveillance platform, along with an active community engagement strategy, guarantees consistent recruitment and retention of participants in clinical trials. The Malaria Diagnostic Center, backed by WHO-certified microscopists and a large malaria blood film collection, champions high-quality malaria diagnosis and strict quality assurance through standardized microscopy trainings. The Malaria Drug Resistance Laboratory leverages cutting-edge technology such as real-time Polymerase Chain Reaction (qPCR) to conduct comprehensive research on resistance markers and obtain information on drug efficacy. The laboratory has been working on validating artemisinin resistance markers and improving tracking methods for current and future antimalarial compounds. Finally, the Basic Science Laboratory employs advanced genomic technology to examine endpoints such as immunogenicity and genomic fingerprinting for candidate drugs and vaccine efficacy. Herein, we examine the site's significant contributions to malaria policy, management, and prevention practices in Kenya and around the world.
Collapse
Affiliation(s)
- Peter M Sifuna
- Kenya Medical Research Institute, Kisumu, Kenya
- U.S. Army Medical Research Directorate-Africa, Kisumu, Kenya
| | - Michal Mbinji
- Kenya Medical Research Institute, Kisumu, Kenya
- U.S. Army Medical Research Directorate-Africa, Kisumu, Kenya
| | - Tina O Lucas
- Kenya Medical Research Institute, Kisumu, Kenya
- U.S. Army Medical Research Directorate-Africa, Kisumu, Kenya
| | - Irene Onyango
- Kenya Medical Research Institute, Kisumu, Kenya
- U.S. Army Medical Research Directorate-Africa, Kisumu, Kenya
| | - Hoseah M Akala
- Kenya Medical Research Institute, Kisumu, Kenya
- U.S. Army Medical Research Directorate-Africa, Kisumu, Kenya
| | - John N Waitumbi
- Kenya Medical Research Institute, Kisumu, Kenya
- U.S. Army Medical Research Directorate-Africa, Kisumu, Kenya
| | - Bernhards R Ogutu
- Kenya Medical Research Institute, Kisumu, Kenya
- U.S. Army Medical Research Directorate-Africa, Kisumu, Kenya
| | - Jack N Hutter
- U.S. Army Medical Research Directorate-Africa, Kisumu, Kenya
| | - Walter Otieno
- Kenya Medical Research Institute, Kisumu, Kenya
- U.S. Army Medical Research Directorate-Africa, Kisumu, Kenya
| |
Collapse
|
19
|
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
|
20
|
Hill J, Bange T, Hoyt J, Kariuki S, Jalloh MF, Webster J, Okello G. Integration of the RTS,S/AS01 malaria vaccine into the Essential Programme on Immunisation in western Kenya: a qualitative longitudinal study from the health system perspective. Lancet Glob Health 2024; 12:e672-e684. [PMID: 38430916 PMCID: PMC10932755 DOI: 10.1016/s2214-109x(24)00013-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Malaria accounts for over half a million child deaths annually. WHO recommends RTS,S/AS01 to prevent malaria in children living in moderate-to-high malaria transmission regions. We conducted a qualitative longitudinal study to investigate the contextual and dynamic factors shaping vaccine delivery and uptake during a pilot introduction in western Kenya. METHODS The study was conducted between Oct 3, 2019, and Mar 24, 2022. We conducted participant and non-participant observations and in-depth interviews with health-care providers, health managers, and national policymakers at three timepoints using an iterative approach and observations of practices and processes of malaria vaccine delivery. Transcripts were coded by content analysis using the consolidated framework for implementation research, to which emerging themes were added deductively and categorised into challenges and opportunities. FINDINGS We conducted 112 in-depth interviews with 60 participants (25 health-care providers, 27 managers, and eight policy makers). Health-care providers highlighted limitations in RTS,S/AS01 integration into routine immunisation services due to the concurrent pilot evaluation and temporary adaptations for health reporting. Initial challenges related to the complexity of the four-dose schedule (up to 24-months); however, self-efficacy increased over time as the health-care providers gained experience in vaccine delivery. Low uptake of the fourth dose remained a challenge. Health managers cited insufficient trained immunisation staff and inadequate funding for supervision. Confidence in the vaccine increased among all participant groups owing to reductions in malaria frequency and severity. INTERPRETATION Integration of RTS,S/AS01 into immunisation services in western Kenya presented substantial operational challenges most of which were overcome in the first 2 years, providing important lessons for other countries. Programme expansion is feasible with intensive staff training and retention, enhanced supervision, and defaulter-tracing to ensure uptake of all doses. FUNDING PATH via World Health Organization; Gavi, the Vaccine Alliance; The Global Fund; and Unitaid.
Collapse
Affiliation(s)
- Jenny Hill
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
| | - Teresa Bange
- Kenya Medical Research Institute/Centre for Global Health Research, Kisumu, Kenya
| | - Jenna Hoyt
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Simon Kariuki
- Kenya Medical Research Institute/Centre for Global Health Research, Kisumu, Kenya
| | - Mohamed F Jalloh
- Global Immunization Division, Global Health Center, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jayne Webster
- Disease Control Department, London School of Tropical Medicine & Hygiene, London, 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
| |
Collapse
|
21
|
Jairoce C, Macià D, Torres-Yaguana JP, Mayer L, Vidal M, Santano R, Hurtado-Guerrero R, Reiter K, Narum DL, Lopez-Gutierrez B, Hamerly T, Sacarlal J, Aguilar R, Dinglasan RR, Moncunill G, Izquierdo L, Dobaño C. RTS,S/AS02A Malaria Vaccine-Induced IgG Responses Equally Recognize Native-Like Fucosylated and Nonfucosylated Plasmodium falciparum Circumsporozoite Proteins. J Infect Dis 2024; 229:795-799. [PMID: 37889513 PMCID: PMC11491836 DOI: 10.1093/infdis/jiad471] [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: 07/06/2023] [Revised: 10/01/2023] [Accepted: 10/26/2023] [Indexed: 10/28/2023] Open
Abstract
The RTS,S/AS02A malaria vaccine is based on the Plasmodium falciparum circumsporozoite protein (PfCSP), which is O-fucosylated on the sporozoite surface. We determined whether RTS,S/AS02A-induced immunoglobulin G (IgG) antibodies recognize vaccine-like nonfucosylated PfCSP better than native-like fucosylated PfCSP. Similar to previous vaccine trials, RTS,S/AS02A vaccination induced high anti-PfCSP IgG levels associated with malaria protection. IgG recognition of nonfucosylated and fucosylated PfCSP was equivalent, suggesting that PfCSP fucosylation does not affect antibody recognition. Clinical Trials Registration. NCT00197041.
Collapse
Affiliation(s)
- Chenjerai Jairoce
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
| | - Dídac Macià
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Jorge P Torres-Yaguana
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Leonie Mayer
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Catalonia, Spain
- Université Claude Bernard Lyon 1, Université de Lyon, Villeurbanne, France
| | - Marta Vidal
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Rebeca Santano
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Ramón Hurtado-Guerrero
- Institute of Biocomputation and Physics of Complex Systems, University of Zaragoza, Zaragoza, Spain
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain
| | - Karine Reiter
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - David L Narum
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Borja Lopez-Gutierrez
- Emerging Pathogens Institute, Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Timothy Hamerly
- Emerging Pathogens Institute, Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Jahit Sacarlal
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
| | - Ruth Aguilar
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Rhoel R Dinglasan
- Emerging Pathogens Institute, Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Gemma Moncunill
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Luis Izquierdo
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Carlota Dobaño
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| |
Collapse
|
22
|
Wang N, Wang T. Innovative translational platforms for rapid developing clinical vaccines against COVID-19 and other infectious disease. Biotechnol J 2024; 19:e2300658. [PMID: 38403469 DOI: 10.1002/biot.202300658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/18/2023] [Accepted: 12/28/2023] [Indexed: 02/27/2024]
Abstract
A vaccine is a biological preparation that contains the antigen capable of stimulating the immune system to form the defense against pathogens. Vaccine development often confronts big challenges, including time/energy-consuming, low efficacy, lag to pathogen emergence and mutation, and even safety concern. However, these seem now mostly conquerable through constructing the advanced translational platforms that can make innovative vaccines, sometimes, potentiated with a distinct multifunctional VADS (vaccine adjuvant delivery system), as evidenced by the development of various vaccines against the covid-19 pandemic at warp speed. Particularly, several covid-19 vaccines, such as the viral-vectored vaccines, mRNA vaccines and DNA vaccines, regarded as the innovative ones that are rapidly made via the high technology-based translational platforms. These products have manifested powerful efficacy while showing no unacceptable safety profile in clinics, allowing them to be approved for massive vaccination at also warp speed. Now, the proprietary translational platforms integrated with the state-of-the-art biotechnologies, and even the artificial intelligence (AI), represent an efficient mode for rapid making innovative clinical vaccines against infections, thus increasingly attracting interests of vaccine research and development. Herein, the advanced translational platforms for making innovative vaccines, together with their design principles and immunostimulatory efficacies, are comprehensively elaborated.
Collapse
Affiliation(s)
- Ning Wang
- School of Food and Biological engineering, Hefei University of Technology, Hefei, Anhui Province, China
| | - Ting Wang
- School of Pharmacy, Anhui Medical University, Hefei, Anhui Province, China
| |
Collapse
|
23
|
Zhu C, Jiao S, Xu W. CD8 + Trms against malaria liver-stage: prospects and challenges. Front Immunol 2024; 15:1344941. [PMID: 38318178 PMCID: PMC10839007 DOI: 10.3389/fimmu.2024.1344941] [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: 11/27/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Attenuated sporozoites provide a valuable model for exploring protective immunity against the malarial liver stage, guiding the design of highly efficient vaccines to prevent malaria infection. Liver tissue-resident CD8+ T cells (CD8+ Trm cells) are considered the host front-line defense against malaria and are crucial to developing prime-trap/target strategies for pre-erythrocytic stage vaccine immunization. However, the spatiotemporal regulatory mechanism of the generation of liver CD8+ Trm cells and their responses to sporozoite challenge, as well as the protective antigens they recognize remain largely unknown. Here, we discuss the knowledge gap regarding liver CD8+ Trm cell formation and the potential strategies to identify predominant protective antigens expressed in the exoerythrocytic stage, which is essential for high-efficacy malaria subunit pre-erythrocytic vaccine designation.
Collapse
Affiliation(s)
- Chengyu Zhu
- The School of Medicine, Chongqing University, Chongqing, China
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shiming Jiao
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wenyue Xu
- The School of Medicine, Chongqing University, Chongqing, China
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
24
|
de Jong HK, Grobusch MP. Monoclonal antibody applications in travel medicine. Trop Dis Travel Med Vaccines 2024; 10:2. [PMID: 38221606 PMCID: PMC10789029 DOI: 10.1186/s40794-023-00212-x] [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: 11/06/2023] [Accepted: 11/21/2023] [Indexed: 01/16/2024] Open
Abstract
For decades, immunoglobulin preparations have been used to prevent or treat infectious diseases. Since only a few years, monoclonal antibody applications (mAbs) are taking flight and are increasingly dominating this field. In 2014, only two mAbs were registered; end of October 2023, more than ten mAbs are registered or have been granted emergency use authorization, and many more are in (pre)clinical phases. Especially the COVID-19 pandemic has generated this surge in licensed monoclonal antibodies, although multiple phase 1 studies were already underway in 2019 for other infectious diseases such as malaria and yellow fever. Monoclonal antibodies could function as prophylaxis (i.e., for the prevention of malaria), or could be used to treat (tropical) infections (i.e., rabies, dengue fever, yellow fever). This review focuses on the discussion of the prospects of, and obstacles for, using mAbs in the prevention and treatment of (tropical) infectious diseases seen in the returning traveler; and provides an update on the mAbs currently being developed for infectious diseases, which could potentially be of interest for travelers.
Collapse
Affiliation(s)
- Hanna K de Jong
- Centre of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Centers, Location AMC, Amsterdam Infection and Immunity, Amsterdam Public Health, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Martin P Grobusch
- Centre of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Centers, Location AMC, Amsterdam Infection and Immunity, Amsterdam Public Health, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Institute of Tropical Medicine & Deutsches Zentrum Für Infektionsforschung, University of Tübingen, Tübingen, Germany
- Centre de Recherches Médicales, (CERMEL), Lambaréné, Gabon
- Masanga Medical Research Unit (MMRU), Masanga, Sierra Leone
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
25
|
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
|
26
|
Mategula D, Gichuki J, Chipeta MG, Chirombo J, Kalonde PK, Gumbo A, Kayange M, Samuel V, Kwizombe C, Hamuza G, Kalanga A, Kamowa D, Mitambo C, Kawonga J, Banda B, Kafulafula J, Banda A, Twabi H, Musa E, Masambuka M, Ntwere T, Ligomba C, Munthali L, Sakala M, Bangoura A, Kapito-Tembo A, Masingi-Mbeye N, Mathanga DP, Terlouw DJ. Two decades of malaria control in Malawi: Geostatistical Analysis of the changing malaria prevalence from 2000-2022. Wellcome Open Res 2024; 8:264. [PMID: 38756913 PMCID: PMC11097645 DOI: 10.12688/wellcomeopenres.19390.2] [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] [Accepted: 05/16/2024] [Indexed: 05/18/2024] Open
Abstract
Background Malaria remains a public health problem in Malawi and has a serious socio-economic impact on the population. In the past two decades, available malaria control measures have been substantially scaled up, such as insecticide-treated bed nets, artemisinin-based combination therapies, and, more recently, the introduction of the malaria vaccine, the RTS,S/AS01. In this paper, we describe the epidemiology of malaria for the last two decades to understand the past transmission and set the scene for the elimination agenda. Methods A collation of parasite prevalence surveys conducted between the years 2000 and 2022 was done. A spatio-temporal geostatistical model was fitted to predict the yearly malaria risk for children aged 2-10 years (PfPR 2-10) at 1×1 km spatial resolutions. Parameter estimation was done using the Monte Carlo maximum likelihood method. District-level prevalence estimates adjusted for population are calculated for the years 2000 to 2022. Results A total of 2,595 sampled unique locations from 2000 to 2022 were identified through the data collation exercise. This represents 70,565 individuals that were sampled in the period. In general, the PfPR2_10 declined over the 22 years. The mean modelled national PfPR2_10 in 2000 was 43.93 % (95% CI:17.9 to 73.8%) and declined to 19.2% (95%CI 7.49 to 37.0%) in 2022. The smoothened estimates of PfPR2_10 indicate that malaria prevalence is very heterogeneous with hotspot areas concentrated on the southern shores of Lake Malawi and the country's central region. Conclusions The last two decades are associated with a decline in malaria prevalence, highly likely associated with the scale-up of control interventions. The country should move towards targeted malaria control approaches informed by surveillance data.
Collapse
Affiliation(s)
- Donnie Mategula
- Malawi-Liverpool Wellcome Programme, Blantyre, Malawi
- Liverpool School of Tropical Medicine, Liverpool, L35QA, UK
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Southern, Malawi
| | - Judy Gichuki
- Strathmore University, Institute of Healthcare Management, Nairobi, Kenya
| | | | | | - Patrick Ken Kalonde
- Malawi-Liverpool Wellcome Programme, Blantyre, Malawi
- Liverpool School of Tropical Medicine, Liverpool, L35QA, UK
| | - Austin Gumbo
- National Malaria Control Programme, Ministry of Health, Lilongwe, Malawi
| | - Michael Kayange
- National Malaria Control Programme, Ministry of Health, Lilongwe, Malawi
| | - Vincent Samuel
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Southern, Malawi
| | - Colins Kwizombe
- U.S. President's Malaria Initiative, United States Agency for International Development (USAID), Lilongwe, Malawi
| | - Gracious Hamuza
- National Malaria Control Programme, Ministry of Health, Lilongwe, Malawi
| | | | - Dina Kamowa
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Southern, Malawi
| | | | - Jacob Kawonga
- Country Health Information Systems and Data Use (CHISU) Program, Lilongwe, Malawi
| | - Benard Banda
- Country Health Information Systems and Data Use (CHISU) Program, Lilongwe, Malawi
| | | | - Akuzike Banda
- National Malaria Control Programme, Ministry of Health, Lilongwe, Malawi
| | - Halima Twabi
- Department of Mathematical Sciences, School of Natural and Applied Sciences, University of Malawi, Zomba, Malawi
| | - Esloyn Musa
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Southern, Malawi
| | | | - Tapiwa Ntwere
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Southern, Malawi
| | | | - Lumbani Munthali
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Southern, Malawi
| | - Melody Sakala
- Malawi-Liverpool Wellcome Programme, Blantyre, Malawi
| | | | - Atupele Kapito-Tembo
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Southern, Malawi
| | - Nyanyiwe Masingi-Mbeye
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Southern, Malawi
| | - Don P. Mathanga
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Southern, Malawi
| | - Dianne J Terlouw
- Malawi-Liverpool Wellcome Programme, Blantyre, Malawi
- Liverpool School of Tropical Medicine, Liverpool, L35QA, UK
| |
Collapse
|
27
|
Scaria PV, Roth N, Schwendt K, Muratova OV, Alani N, Lambert LE, Barnafo EK, Rowe CG, Zaidi IU, Rausch KM, Narum DL, Petsch B, Duffy PE. mRNA vaccines expressing malaria transmission-blocking antigens Pfs25 and Pfs230D1 induce a functional immune response. NPJ Vaccines 2024; 9:9. [PMID: 38184666 PMCID: PMC10771442 DOI: 10.1038/s41541-023-00783-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/22/2023] [Indexed: 01/08/2024] Open
Abstract
Malaria transmission-blocking vaccines (TBV) are designed to inhibit the sexual stage development of the parasite in the mosquito host and can play a significant role in achieving the goal of malaria elimination. Preclinical and clinical studies using protein-protein conjugates of leading TBV antigens Pfs25 and Pfs230 domain 1 (Pfs230D1) have demonstrated the feasibility of TBV. Nevertheless, other promising vaccine platforms for TBV remain underexplored. The recent success of mRNA vaccines revealed the potential of this technology for infectious diseases. We explored the mRNA platform for TBV development. mRNA constructs of Pfs25 and Pfs230D1 variously incorporating signal peptides (SP), GPI anchor, and Trans Membrane (TM) domain were assessed in vitro for antigen expression, and selected constructs were evaluated in mice. Only mRNA constructs with GPI anchor or TM domain that resulted in high cell surface expression of the antigens yielded strong immune responses in mice. These mRNA constructs generated higher transmission-reducing functional activity versus the corresponding alum-adjuvanted protein-protein conjugates used as comparators. Pfs25 mRNA with GPI anchor or TM maintained >99% transmission reducing activity through 126 days, the duration of the study, demonstrating the potential of mRNA platform for TBV.
Collapse
Affiliation(s)
- Puthupparampil V Scaria
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | | | | | - Olga V Muratova
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | - Nada Alani
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | - Lynn E Lambert
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | - Emma K Barnafo
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | - Christopher G Rowe
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | - Irfan U Zaidi
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | - Kelly M Rausch
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | - David L Narum
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA
| | | | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, 29 Lincoln Drive, Building 29B, MD, 20892-2903, USA.
| |
Collapse
|
28
|
Roman F, Burny W, Ceregido MA, Laupèze B, Temmerman ST, Warter L, Coccia M. Adjuvant system AS01: from mode of action to effective vaccines. Expert Rev Vaccines 2024; 23:715-729. [PMID: 39042099 DOI: 10.1080/14760584.2024.2382725] [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: 04/17/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION The use of novel adjuvants in human vaccines continues to expand as their contribution to preventing disease in challenging populations and caused by complex pathogens is increasingly understood. AS01 is a family of liposome-based vaccine Adjuvant Systems containing two immunostimulants: 3-O-desacyl-4'-monophosphoryl lipid A and the saponin QS-21. AS01-containing vaccines have been approved and administered to millions of individuals worldwide. AREAS COVERED Here, we report advances in our understanding of the mode of action of AS01 that contributed to the development of efficacious vaccines preventing disease due to malaria, herpes zoster, and respiratory syncytial virus. AS01 induces early innate immune activation that induces T cell-mediated and antibody-mediated responses with optimized functional characteristics and induction of immune memory. AS01-containing vaccines appear relatively impervious to baseline immune status translating into high efficacy across populations. Currently licensed AS01-containing vaccines have shown acceptable safety profiles in clinical trials and post-marketing settings. EXPERT OPINION Initial expectations that adjuvantation with AS01 could support effective vaccine responses and contribute to disease control have been realized. Investigation of the utility of AS01 in vaccines to prevent other challenging diseases, such as tuberculosis, is ongoing, together with efforts to fully define its mechanisms of action in different vaccine settings.
Collapse
|
29
|
Poria R, Kala D, Nagraik R, Dhir Y, Dhir S, Singh B, Kaushik NK, Noorani MS, Kaushal A, Gupta S. Vaccine development: Current trends and technologies. Life Sci 2024; 336:122331. [PMID: 38070863 DOI: 10.1016/j.lfs.2023.122331] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/24/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023]
Abstract
Despite the effectiveness of vaccination in reducing or eradicating diseases caused by pathogens, there remain certain diseases and emerging infections for which developing effective vaccines is inherently challenging. Additionally, developing vaccines for individuals with compromised immune systems or underlying medical conditions presents significant difficulties. As well as traditional vaccine different methods such as inactivated or live attenuated vaccines, viral vector vaccines, and subunit vaccines, emerging non-viral vaccine technologies, including viral-like particle and nanoparticle vaccines, DNA/RNA vaccines, and rational vaccine design, offer new strategies to address the existing challenges in vaccine development. These advancements have also greatly enhanced our understanding of vaccine immunology, which will guide future vaccine development for a broad range of diseases, including rapidly emerging infectious diseases like COVID-19 and diseases that have historically proven resistant to vaccination. This review provides a comprehensive assessment of emerging non-viral vaccine production methods and their application in addressing the fundamental and current challenges in vaccine development.
Collapse
Affiliation(s)
- Renu Poria
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India
| | - Deepak Kala
- Centera Laboratories, Institute of High Pressure Physics PAS, 01-142 Warsaw, Poland
| | - Rupak Nagraik
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh, India
| | - Yashika Dhir
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India
| | - Sunny Dhir
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India
| | - Bharat Singh
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India
| | - Naveen Kumar Kaushik
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, Sector-125, Noida, Uttar Pradesh, India
| | - Md Salik Noorani
- Department of Botany, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Ankur Kaushal
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India.
| | - Shagun Gupta
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India.
| |
Collapse
|
30
|
Price J, Gurley N, Gyapong M, Ansah EK, Awusabo-Asare K, Gyasi SF, Nkhoma P, Nyondo-Mipando AL, Okello G, Webster J, Desmond N, Hill J, Gordon WS. Acceptance of and Adherence to a Four-Dose RTS,S/AS01 Schedule: Findings from a Longitudinal Qualitative Evaluation Study for the Malaria Vaccine Implementation Programme. Vaccines (Basel) 2023; 11:1801. [PMID: 38140205 PMCID: PMC10747521 DOI: 10.3390/vaccines11121801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/19/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND The WHO recommended the use of the RTS,S/AS01 malaria vaccine (RTS,S) based on a pilot evaluation in routine use in Ghana, Kenya, and Malawi. A longitudinal qualitative study was conducted to examine facilitators and barriers to uptake of a 4-dose RTS,S schedule. METHODS A cohort of 198 caregivers of RTS,S-eligible children from communities where RTS,S was provided through the pilot were interviewed three times over a ≈22-month, 4-dose schedule. The interviews examined caregiver perceptions and behaviors. Children's vaccination history was obtained to determine dose uptake. RESULTS 162 caregivers remained at round 3 (R3); vaccination history was available for 152/162 children. Despite early rumors/fears, the uptake of initial doses was high, driven by vaccine trust. Fears dissipated by R2, replaced with an enthusiasm for RTS,S as caregivers perceived its safety and less frequent and severe malaria. By R3, 98/152 children had received four doses; 34 three doses; 9 one or two doses; and 11 zero doses. The health system and information barriers were important across all under-dose cases. Fears about AEFIs/safety were important in zero-, one-, and two-dose cases. Competing life/livelihood demands and complacency were found in three-dose cases. Regardless of the doses received, caregivers had positive attitudes towards RTS,S by R3. CONCLUSIONS Findings from our study will help countries newly introducing the vaccine to anticipate and preempt reasons for delayed acceptance and missed RTS,S doses.
Collapse
Affiliation(s)
| | - Nikki Gurley
- King County Department of Community and Human Services, 401 5th Ave #500, Seattle, WA 98104, USA;
| | - Margaret Gyapong
- Institute of Health Research, University of Health and Allied Sciences, PMB 31, Ho, Volta Region, Ghana;
| | - Evelyn Korkor Ansah
- Center for Malaria Research, University of Health and Allied Sciences, PMB 31, Ho, Volta Region, Ghana;
| | - Kofi Awusabo-Asare
- Department of Population and Health, University of Cape Coast, New Administration Block, Cape Coast, Ghana;
| | - Samuel Fosu Gyasi
- Center for Research in Applied Biology, School of Sciences, University of Energy and Natural Resources, P.O. Box 214, Sunyani, Ghana;
| | - Pearson Nkhoma
- Goldsmiths, University of London, 8 Lewisham Way, New Cross, London SE14 6NW, UK;
| | - Alinane Linda Nyondo-Mipando
- Department of Health Systems and Policy, Kamuzu University of Health Sciences, Private Bag 360, Blantyre 3, Malawi;
| | - George Okello
- Kenya Medical Research Institute, Centre for Geographic Medicine, Kisumu P.O. Box 1578, Kenya;
| | - Jayne Webster
- London School of Hygiene and Tropical Medicine, Keppel St, London WC1E 7HT, UK;
| | - Nicola Desmond
- Department of International Public Health, Liverpool School of Tropical Medicine, University of Liverpool, Pembroke Pl, Liverpool L3 5QA, UK;
| | - Jenny Hill
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Pembroke Pl, Liverpool L3 5QA, UK;
| | | |
Collapse
|
31
|
Powell TJ, Tang J, Mitchell R, DeRome ME, Jacobs A, Palath N, Cardenas E, Yorke M, Boyd JG, Kaba SA, Nardin E. Immunogenicity, Efficacy, and Safety of a Novel Synthetic Microparticle Pre-Erythrocytic Malaria Vaccine in Multiple Host Species. Vaccines (Basel) 2023; 11:1789. [PMID: 38140193 PMCID: PMC10748200 DOI: 10.3390/vaccines11121789] [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: 10/23/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
We previously reported a protective antibody response in mice immunized with synthetic microparticle vaccines made using layer-by-layer fabrication (LbL-MP) and containing the conserved T1BT* epitopes from the P. falciparum circumsporozoite protein. To further optimize the vaccine candidate, a benchtop tangential flow filtration method (LbL-by-TFF) was developed and utilized to produce vaccine candidates that differed in the status of base layer crosslinking, inclusion of a TLR2 ligand in the antigenic peptide, and substitution of serine or alanine for an unpaired cysteine residue in the T* epitope. Studies in mice revealed consistent superiority of the Pam3Cys-modified candidates and a modest benefit of base layer crosslinking, as evidenced by higher and more persistent antibody titers (up to 18 months post-immunization), a qualitative improvement of T-cell responses toward a Th1 phenotype, and greater protection from live parasite challenges compared to the unmodified prototype candidate. Immunogenicity was also tested in a non-human primate model, the rhesus macaque. Base layer-crosslinked LbL-MP loaded with T1BT* peptide with or without covalently linked Pam3Cys elicited T1B-specific antibody responses and T1BT*-specific T-cell responses dominated by IFNγ secretion with lower levels of IL-5 secretion. The Pam3Cys-modified construct was more potent, generating antibody responses that neutralized wild-type P. falciparum in an in vitro hepatocyte invasion assay. IgG purified from individual macaques immunized with Pam3Cys.T1BT* LbL-MP protected naïve mice from challenges with transgenic P. berghei sporozoites that expressed the full-length PfCS protein, with 50-88% of passively immunized mice parasite-free for ≥15 days. Substitution of serine for an unpaired cysteine in the T* region of the T1BT* subunit did not adversely impact immune potency in the mouse while simplifying the manufacture of the antigenic peptide. In a Good Laboratory Practices compliant rabbit toxicology study, the base layer-crosslinked, Pam3Cys-modified, serine-substituted candidate was shown to be safe and immunogenic, eliciting parasite-neutralizing antibody responses and establishing the dose/route/regimen for a clinical evaluation of this novel synthetic microparticle pre-erythrocytic malaria vaccine candidate.
Collapse
Affiliation(s)
- Thomas J. Powell
- Artificial Cell Technologies, Inc., 5 Science Park, Suite 13, New Haven, CT 06511, USA; (J.T.); (M.E.D.); (A.J.); (N.P.); (E.C.); (M.Y.); (J.G.B.)
| | - Jie Tang
- Artificial Cell Technologies, Inc., 5 Science Park, Suite 13, New Haven, CT 06511, USA; (J.T.); (M.E.D.); (A.J.); (N.P.); (E.C.); (M.Y.); (J.G.B.)
| | - Robert Mitchell
- Department of Microbiology, School of Medicine, New York University, New York, NY 10010, USA; (R.M.); (E.N.)
| | - Mary E. DeRome
- Artificial Cell Technologies, Inc., 5 Science Park, Suite 13, New Haven, CT 06511, USA; (J.T.); (M.E.D.); (A.J.); (N.P.); (E.C.); (M.Y.); (J.G.B.)
- Multiple Myeloma Research Foundation, 383 Main Avenue, 5th Floor, Norwalk, CT 06851, USA
| | - Andrea Jacobs
- Artificial Cell Technologies, Inc., 5 Science Park, Suite 13, New Haven, CT 06511, USA; (J.T.); (M.E.D.); (A.J.); (N.P.); (E.C.); (M.Y.); (J.G.B.)
| | - Naveen Palath
- Artificial Cell Technologies, Inc., 5 Science Park, Suite 13, New Haven, CT 06511, USA; (J.T.); (M.E.D.); (A.J.); (N.P.); (E.C.); (M.Y.); (J.G.B.)
- Pfizer, Inc., Andover, MA 01810, USA
| | - Edwin Cardenas
- Artificial Cell Technologies, Inc., 5 Science Park, Suite 13, New Haven, CT 06511, USA; (J.T.); (M.E.D.); (A.J.); (N.P.); (E.C.); (M.Y.); (J.G.B.)
| | - Michelle Yorke
- Artificial Cell Technologies, Inc., 5 Science Park, Suite 13, New Haven, CT 06511, USA; (J.T.); (M.E.D.); (A.J.); (N.P.); (E.C.); (M.Y.); (J.G.B.)
| | - James G. Boyd
- Artificial Cell Technologies, Inc., 5 Science Park, Suite 13, New Haven, CT 06511, USA; (J.T.); (M.E.D.); (A.J.); (N.P.); (E.C.); (M.Y.); (J.G.B.)
| | - Stephen A. Kaba
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA;
- GreenLight Biosciences, Inc., Lexington, MA 02421, USA
| | - Elizabeth Nardin
- Department of Microbiology, School of Medicine, New York University, New York, NY 10010, USA; (R.M.); (E.N.)
| |
Collapse
|
32
|
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
|
33
|
Mellid-Carballal R, Gutierrez-Gutierrez S, Rivas C, Garcia-Fuentes M. Viral protein-based nanoparticles (part 2): Pharmaceutical applications. Eur J Pharm Sci 2023; 189:106558. [PMID: 37567394 DOI: 10.1016/j.ejps.2023.106558] [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: 02/17/2023] [Revised: 07/10/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
Viral protein nanoparticles (ViP NPs) such as virus-like particles and virosomes are structures halfway between viruses and synthetic nanoparticles. The biological nature of ViP NPs endows them with the biocompatibility, biodegradability, and functional properties that many synthetic nanoparticles lack. At the same time, the absence of a viral genome avoids the safety concerns of viruses. Such characteristics of ViP NPs offer a myriad of opportunities for theirapplication at several points across disease development: from prophylaxis to diagnosis and treatment. ViP NPs present remarkable immunostimulant properties, and thus the vaccination field has benefited the most from these platforms capable of overcoming the limitations of both traditional and subunit vaccines. This was reflected in the marketing authorization of several VLP- and virosome-based vaccines. Besides, ViP NPs inherit the ability of viruses to deliver their cargo to target cells. Because of that, ViP NPs are promising candidates as vectors for drug and gene delivery, and for diagnostic applications. In this review, we analyze the pharmaceutical applications of ViP NPs, describing the products that are commercially available or under clinical evaluation, but also the advances that scientists are making toward the implementation of ViP NPs in other areas of major pharmaceutical interest.
Collapse
Affiliation(s)
- Rocio Mellid-Carballal
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain
| | - Sara Gutierrez-Gutierrez
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain
| | - Carmen Rivas
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Universidad de Santiago de Compostela, Spain; Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología (CNB)-CSIC, Spain
| | - Marcos Garcia-Fuentes
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Universidad de Santiago de Compostela, Spain.
| |
Collapse
|
34
|
Bechtold P, Wagner P, Hosch S, Gregorini M, Stark WJ, Gody JC, Kodia-Lenguetama ER, Pagonendji MS, Donfack OT, Phiri WP, García GA, Nsanzanbana C, Daubenberger CA, Schindler T, Vickos U. Development and evaluation of PlasmoPod: A cartridge-based nucleic acid amplification test for rapid malaria diagnosis and surveillance. PLOS GLOBAL PUBLIC HEALTH 2023; 3:e0001516. [PMID: 37756280 PMCID: PMC10529553 DOI: 10.1371/journal.pgph.0001516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 09/04/2023] [Indexed: 09/29/2023]
Abstract
Malaria surveillance is hampered by the widespread use of diagnostic tests with low sensitivity. Adequate molecular malaria diagnostics are often only available in centralized laboratories. PlasmoPod is a novel cartridge-based nucleic acid amplification test for rapid, sensitive, and quantitative detection of malaria parasites. PlasmoPod is based on reverse-transcription quantitative polymerase chain reaction (RT-qPCR) of the highly abundant Plasmodium spp. 18S ribosomal RNA/DNA biomarker and is run on a portable qPCR instrument which allows diagnosis in less than 30 minutes. Our analytical performance evaluation indicates that a limit-of-detection as low as 0.02 parasites/μL can be achieved and no cross-reactivity with other pathogens common in malaria endemic regions was observed. In a cohort of 102 asymptomatic individuals from Bioko Island with low malaria parasite densities, PlasmoPod accurately detected 83 cases, resulting in an overall detection rate of 81.4%. Notably, there was a strong correlation between the Cq values obtained from the reference RT-qPCR assay and those obtained from PlasmoPod. In an independent cohort, using dried blood spots from malaria symptomatic children living in the Central African Republic, we demonstrated that PlasmoPod outperforms malaria rapid diagnostic tests based on the PfHRP2 and panLDH antigens as well as thick blood smear microscopy. Our data suggest that this 30-minute sample-to-result RT-qPCR procedure is likely to achieve a diagnostic performance comparable to a standard laboratory-based RT-qPCR setup. We believe that the PlasmoPod rapid NAAT could enable widespread accessibility of high-quality and cost-effective molecular malaria surveillance data through decentralization of testing and surveillance activities, especially in elimination settings.
Collapse
Affiliation(s)
- Philippe Bechtold
- Institute for Chemical and Bioengineering, ETH Zurich, Zuerich, Switzerland
- Diaxxo AG, Zuerich, Switzerland
| | - Philipp Wagner
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Salome Hosch
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Michele Gregorini
- Institute for Chemical and Bioengineering, ETH Zurich, Zuerich, Switzerland
- Diaxxo AG, Zuerich, Switzerland
| | - Wendelin J. Stark
- Institute for Chemical and Bioengineering, ETH Zurich, Zuerich, Switzerland
- Diaxxo AG, Zuerich, Switzerland
| | - Jean Chrysostome Gody
- Paediatric Hospital and University Complex of Bangui, Bangui, Central African Republic
| | | | | | | | | | | | - Christian Nsanzanbana
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Claudia A. Daubenberger
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Tobias Schindler
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Ulrich Vickos
- Infectious and Tropical Diseases Unit, Department of Medicine, Amitié Hospital, Bangui, Central African Republic
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| |
Collapse
|
35
|
Alonso A, Alcolea PJ, Larraga J, Peris MP, Esteban A, Cortés A, Ruiz-García S, Castillo JA, Larraga V. A non-replicative antibiotic resistance-free DNA vaccine delivered by the intranasal route protects against canine leishmaniasis. Front Immunol 2023; 14:1213193. [PMID: 37790927 PMCID: PMC10543895 DOI: 10.3389/fimmu.2023.1213193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/11/2023] [Indexed: 10/05/2023] Open
Abstract
Leishmania infantum is the etiological agent of zoonotic visceral leishmaniasis (ZVL). The disease is endemic in Central and South America, Central and South East Asia, and the Mediterranean basin. Dogs are the main reservoir, with an estimated prevalence of approximately 2.5 million dogs in Southern Europe. Current treatments cause side effects, disease recurrence, and drug resistance. Therefore, the development of vaccines against canine leishmaniasis is necessary. We have generated a DNA vaccine based on the non-replicative antibiotic resistance marker-free plasmid vector pPAL that contains the encoding gene for the L. infantum activated protein kinase C receptor analog (LACK). Homologous pPAL-LACK prime-boost intranasal administration confers efficacious protection in Beagle dogs with a reduction of clinical signs and a statistically significant reduction of the parasite burden in the bone marrow of more than 90% of dogs after experimental infection with highly infective promastigotes. This DNA vaccine elicits a robust cellular immune response skewed towards the Th1 profile.
Collapse
Affiliation(s)
- Ana Alonso
- Laboratory of Molecular Parasitology and Vaccines, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CIBMS-CSIC), Madrid, Spain
| | - Pedro José Alcolea
- Laboratory of Molecular Parasitology and Vaccines, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CIBMS-CSIC), Madrid, Spain
| | - Jaime Larraga
- Laboratory of Molecular Parasitology and Vaccines, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CIBMS-CSIC), Madrid, Spain
| | - María Paz Peris
- Laboratory of Parasitology, Department of Animal Pathology, Faculty of Veterinary Science, University of Zaragoza, Zaragoza, Spain
| | - Adriana Esteban
- Laboratory of Parasitology, Department of Animal Pathology, Faculty of Veterinary Science, University of Zaragoza, Zaragoza, Spain
| | - Alberto Cortés
- Laboratory of Parasitology, Department of Animal Pathology, Faculty of Veterinary Science, University of Zaragoza, Zaragoza, Spain
| | - Silvia Ruiz-García
- Laboratory of Molecular Parasitology and Vaccines, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CIBMS-CSIC), Madrid, Spain
| | - Juan Antonio Castillo
- Laboratory of Parasitology, Department of Animal Pathology, Faculty of Veterinary Science, University of Zaragoza, Zaragoza, Spain
| | - Vicente Larraga
- Laboratory of Molecular Parasitology and Vaccines, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CIBMS-CSIC), Madrid, Spain
| |
Collapse
|
36
|
Nyandele JP, Kibondo UA, Issa F, Van Geertruyden JP, Warimwe G, Jongo S, Abdulla S, Olotu A. Pre-vaccination monocyte-to-lymphocyte ratio as a biomarker for the efficacy of malaria candidate vaccines: A subgroup analysis of pooled clinical trial data. PLoS One 2023; 18:e0291244. [PMID: 37708143 PMCID: PMC10501550 DOI: 10.1371/journal.pone.0291244] [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: 06/23/2022] [Accepted: 08/24/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND Pre-vaccination monocyte-to-lymphocyte ratio was previously suggested as a marker for malaria vaccine effectiveness. We investigated the potential of this cell ratio as a marker for malaria vaccine efficacy and effectiveness. Effectiveness was investigated by using clinical malaria endpoint, and efficacy was investigated by using surrogate endpoints of Plasmodium falciparum prepatent period, parasite density, and multiplication rates in a controlled human malaria infection trial (CHMI). METHODS We evaluated the correlation between monocyte-to-lymphocyte ratio and RTS,S vaccine effectiveness using Cox regression modeling with clinical malaria as the primary endpoint. Of the 1704 participants in the RTS,S field trial, data on monocyte-to-lymphocyte ratio was available for 842 participants, of whom our analyses were restricted. We further used Spearman Correlations and Cox regression modeling to evaluate the correlation between monocyte-to-lymphocyte ratio and Whole Sporozoite malaria vaccine efficacy using the surrogate endpoints. Of the 97 participants in the controlled human malaria infection vaccine trials, hematology and parasitology information were available for 82 participants, of whom our analyses were restricted. RESULTS The unadjusted efficacy of RTS,S malaria vaccine was 54% (95% CI: 37%-66%, p <0.001). No correlation was observed between monocyte-to-lymphocyte ratio and RTS,S vaccine efficacy (Hazard Rate (HR):0.90, 95%CI:0.45-1.80; p = 0.77). The unadjusted efficacy of Whole Sporozoite malaria vaccine in the appended dataset was 17.6% (95%CI:10%-28.5%, p<0.001). No association between monocyte-to-lymphocyte ratio and the Whole Sporozoite malaria vaccine was found against either the prepatent period (HR = 1.16; 95%CI:0.51-2.62, p = 0.72), parasite density (rho = 0.004, p = 0.97) or multiplication rates (rho = 0.031, p = 0.80). CONCLUSION Monocyte-to-lymphocyte ratio alone may not be an adequate marker for malaria vaccine efficacy. Further investigations on immune correlates and underlying mechanisms of immune protection against malaria could provide a clearer explanation of the differences between those protected in comparison with those not protected against malaria by vaccination.
Collapse
Affiliation(s)
- Jane Paula Nyandele
- Global Health Institute, University of Antwerp, Antwerp, Belgium
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | - Ummi Abdul Kibondo
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | - Fatuma Issa
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | | | | | - Said Jongo
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | - Salim Abdulla
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | - Ally Olotu
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| |
Collapse
|
37
|
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: 1] [Impact Index Per Article: 1.0] [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
|
38
|
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: 3.0] [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
|
39
|
Shukla N, Tang WK, Coelho CH, Long CA, Healy SA, Sagara I, Miura K, Duffy PE, Tolia NH. A human antibody epitope map of the malaria vaccine antigen Pfs25. NPJ Vaccines 2023; 8:108. [PMID: 37542029 PMCID: PMC10403551 DOI: 10.1038/s41541-023-00712-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 07/12/2023] [Indexed: 08/06/2023] Open
Abstract
Pfs25 is a leading antigen for a malaria transmission-blocking vaccine and shows moderate transmission-blocking activity and induction of rapidly decreasing antibody titers in clinical trials. A comprehensive definition of all transmission-reducing epitopes of Pfs25 will inform structure-guided design to enhance Pfs25-based vaccines, leading to potent transmission-blocking activity. Here, we compiled a detailed human antibody epitope map comprising epitope binning data and structures of multiple human monoclonal antibodies, including three new crystal structures of Pfs25 in complex with transmission-reducing antibodies from Malian volunteers immunized with Pfs25 conjugated to EPA and adjuvanted with AS01. These structures revealed additional epitopes in Pfs25 capable of reducing transmission and expanded this characterization to malaria-exposed humans. This work informs immunogen design to focus the antibody response to transmission-reducing epitopes of Pfs25, enabling development of more potent transmission-blocking vaccines for malaria.
Collapse
Affiliation(s)
- Niharika Shukla
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Wai Kwan Tang
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Camila H Coelho
- Pathogenesis and Immunity Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Sara A Healy
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Issaka Sagara
- Malaria Research and Training Center, University of Sciences, Techniques, and Technology, Bamako, Mali
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Patrick E Duffy
- Pathogenesis and Immunity Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Niraj H Tolia
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA.
| |
Collapse
|
40
|
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: 7.0] [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
|
41
|
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: 1.0] [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
|
42
|
González-Sanz M, Berzosa P, Norman FF. Updates on Malaria Epidemiology and Prevention Strategies. Curr Infect Dis Rep 2023; 25:1-9. [PMID: 37361492 PMCID: PMC10248987 DOI: 10.1007/s11908-023-00805-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2023] [Indexed: 06/28/2023]
Abstract
Purpose of Review The objective of this review was to provide an update on recent malaria epidemiology, both globally and in non-endemic areas, to identify the current distribution and repercussions of genetically diverse Plasmodium species and summarize recently implemented intervention and prevention tools. Recent Findings Notable changes in malaria epidemiology have occurred in recent years, with an increase in the number of total cases and deaths globally during 2020-2021, in part attributed to the COVID-19 pandemic. The emergence of artemisinin-resistant species in new areas and the expanding distribution of parasites harbouring deletions of the pfhrp2/3 genes have been concerning. New strategies to curb the burden of this infection, such as vaccination, have been implemented in certain endemic areas and their performance is currently being evaluated. Summary Inadequate control of malaria in endemic regions may have an effect on imported malaria and measures to prevent re-establishment of transmission in malaria-free areas are essential. Enhanced surveillance and investigation of Plasmodium spp. genetic variations will contribute to the successful diagnosis and treatment of malaria in future. Novel strategies for an integrated One Health approach to malaria control should also be strengthened.
Collapse
Affiliation(s)
- Marta González-Sanz
- Infectious Diseases Department, National Referral Unit for Tropical Diseases, Ramón y Cajal University Hospital, IRYCIS, Universidad de Alcalá, CIBER de Enfermedades Infecciosas, Madrid, Spain
| | - Pedro Berzosa
- Malaria and Neglected Tropical Diseases Laboratory, National Centre for Tropical Medicine, Carlos III Health Institute, CIBER de Enfermedades Infecciosas, Madrid, Spain
| | - Francesca F. Norman
- Infectious Diseases Department, National Referral Unit for Tropical Diseases, Ramón y Cajal University Hospital, IRYCIS, Universidad de Alcalá, CIBER de Enfermedades Infecciosas, Madrid, Spain
| |
Collapse
|
43
|
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: 7.0] [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
|
44
|
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
|
45
|
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: 16] [Impact Index Per Article: 16.0] [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
|
46
|
Tripathi P, Bender MF, Lei H, Da Silva Pereira L, Shen CH, Bonilla B, Dillon M, Ou L, Pancera M, Wang LT, Zhang B, Batista FD, Idris AH, Seder RA, Kwong PD. Cryo-EM structures of anti-malarial antibody L9 with circumsporozoite protein reveal trimeric L9 association and complete 27-residue epitope. Structure 2023; 31:480-491.e4. [PMID: 36931276 PMCID: PMC10237622 DOI: 10.1016/j.str.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/17/2023] [Accepted: 02/13/2023] [Indexed: 03/18/2023]
Abstract
Monoclonal antibody L9 recognizes the Plasmodium falciparum circumsporozoite protein (PfCSP) and is highly protective following controlled human malaria challenge. To gain insight into its function, we determined cryoelectron microscopy (cryo-EM) structures of L9 in complex with full-length PfCSP and assessed how this recognition influenced protection by wild-type and mutant L9s. Cryo-EM reconstructions at 3.6- and 3.7-Å resolution revealed L9 to recognize PfCSP as an atypical trimer. Each of the three L9s in the trimer directly recognized an Asn-Pro-Asn-Val (NPNV) tetrapeptide on PfCSP and interacted homotypically to facilitate L9-trimer assembly. We analyzed peptides containing different repeat tetrapeptides for binding to wild-type and mutant L9s to delineate epitope and homotypic components of L9 recognition; we found both components necessary for potent malaria protection. Last, we found the 27-residue stretch recognized by L9 to be highly conserved in P. falciparum isolates, suggesting the newly revealed complete L9 epitope to be an attractive vaccine target.
Collapse
Affiliation(s)
- Prabhanshu Tripathi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael F Bender
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Haotian Lei
- Research Technology Branch Electron Microscopy Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lais Da Silva Pereira
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chen-Hsiang Shen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brian Bonilla
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marlon Dillon
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Li Ou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marie Pancera
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Lawrence T Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Baoshan Zhang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Facundo D Batista
- Departments of Immunology and Microbiology, Harvard Medical School, Boston, MA 02139, USA
| | - Azza H Idris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
47
|
Siddiqui AJ, Bhardwaj J, Saxena J, Jahan S, Snoussi M, Bardakci F, Badraoui R, Adnan M. A Critical Review on Human Malaria and Schistosomiasis Vaccines: Current State, Recent Advancements, and Developments. Vaccines (Basel) 2023; 11:vaccines11040792. [PMID: 37112704 PMCID: PMC10146311 DOI: 10.3390/vaccines11040792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/07/2023] Open
Abstract
Malaria and schistosomiasis are two major parasitic diseases that remain leading causes of morbidity and mortality worldwide. Co-infections of these two parasites are common in the tropics, where both diseases are endemic. The clinical consequences of schistosomiasis and malaria are determined by a variety of host, parasitic, and environmental variables. Chronic schistosomiasis causes malnutrition and cognitive impairments in children, while malaria can cause fatal acute infections. There are effective drugs available to treat malaria and schistosomiasis. However, the occurrence of allelic polymorphisms and the rapid selection of parasites with genetic mutations can confer reduced susceptibility and lead to the emergence of drug resistance. Moreover, the successful elimination and complete management of these parasites are difficult due to the lack of effective vaccines against Plasmodium and Schistosoma infections. Therefore, it is important to highlight all current vaccine candidates undergoing clinical trials, such as pre-erythrocytic and erythrocytic stage malaria, as well as a next-generation RTS,S-like vaccine, the R21/Matrix-M vaccine, that conferred 77% protection against clinical malaria in a Phase 2b trial. Moreover, this review also discusses the progress and development of schistosomiasis vaccines. Furthermore, significant information is provided through this review on the effectiveness and progress of schistosomiasis vaccines currently under clinical trials, such as Sh28GST, Sm-14, and Sm-p80. Overall, this review provides insights into recent progress in malarial and schistosomiasis vaccines and their developmental approaches.
Collapse
Affiliation(s)
- Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
| | - Jyoti Bhardwaj
- Division of Infectious Diseases, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Juhi Saxena
- Department of Biotechnology, University Institute of Biotechnology, Chandigarh University, Gharuan, NH-95, Ludhiana—Chandigarh State Hwy, Mohali 140413, India
| | - Sadaf Jahan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia
| | - Mejdi Snoussi
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
- Laboratory of Genetics, Biodiversity and Valorization of Bio-Resources (LR11ES41), Higher Institute of Biotechnology of Monastir, University of Monastir, Avenue TaharHaddas BP74, Monastir 5000, Tunisia
| | - Fevzi Bardakci
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
| | - Riadh Badraoui
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
- Section of Histology-Cytology, Medicine Faculty of Tunis, University of Tunis El Manar, Tunis 1017, Tunisia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
| |
Collapse
|
48
|
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
|
49
|
Dieng CC, Ford CT, Lerch A, Doniou D, Vegesna K, Janies D, Cui L, Amoah L, Afrane Y, Lo E. Genetic variations of Plasmodium falciparum circumsporozoite protein and the impact on interactions with human immunoproteins and malaria vaccine efficacy. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023; 110:105418. [PMID: 36841398 DOI: 10.1016/j.meegid.2023.105418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 02/27/2023]
Abstract
In October 2021, the world's first malaria vaccine RTS,S was endorsed by WHO for broad use in children, despite its low efficacy. This study examined polyclonal infections and the associations of parasite genetic variations with binding affinity to human leukocyte antigen (HLA). Multiplicity of infection was determined by amplicon deep sequencing of PfMSP1. Genetic variations in PfCSP were examined across 88 samples from Ghana and analyzed together with 1655 PfCSP sequences from other African and non-African isolates. Binding interactions of PfCSP peptide variants and HLA were predicted using NetChop and HADDOCK. High polyclonality was detected among infections, with each infection harboring multiple non-3D7 PfCSP variants. Twenty-seven PfCSP haplotypes were detected in the Ghanaian samples, and they broadly represented PfCSP diversity across Africa. The number of genetic differences between 3D7 and non-3D7 PfCSP variants does not influence binding to HLA. However, CSP peptide length after proteolytic degradation significantly affects its molecular weight and binding affinity to HLA. Despite the high diversity of HLA, the majority of the HLAI and II alleles interacted/bound with all Ghana CSP peptides. Multiple non-3D7 strains among P. falciparum infections could impact the effectiveness of RTS,S. Longer peptides of the Th2R/Th3R CSP regions should be considered in future versions of RTS,S.
Collapse
Affiliation(s)
- Cheikh Cambel Dieng
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA.
| | - Colby T Ford
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA; School of Data Science, University of North Carolina at Charlotte, Charlotte, NC, USA.
| | - Anita Lerch
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Dickson Doniou
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Kovidh Vegesna
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Daniel Janies
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Linda Amoah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana; West Africa Center for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
| | - Yaw Afrane
- Department of Microbiology, University of Ghana Medical School, University of Ghana, Accra, Ghana
| | - Eugenia Lo
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA; School of Data Science, University of North Carolina at Charlotte, Charlotte, NC, USA.
| |
Collapse
|
50
|
Frost I, Sati H, Garcia-Vello P, Hasso-Agopsowicz M, Lienhardt C, Gigante V, Beyer P. The role of bacterial vaccines in the fight against antimicrobial resistance: an analysis of the preclinical and clinical development pipeline. THE LANCET. MICROBE 2023; 4:e113-e125. [PMID: 36528040 PMCID: PMC9892012 DOI: 10.1016/s2666-5247(22)00303-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/10/2022] [Accepted: 10/13/2022] [Indexed: 12/23/2022]
Abstract
Vaccines can be highly effective tools in combating antimicrobial resistance as they reduce infections caused by antibiotic-resistant bacteria and antibiotic consumption associated with disease. This Review looks at vaccine candidates that are in development against pathogens on the 2017 WHO bacterial priority pathogen list, in addition to Clostridioides difficile and Mycobacterium tuberculosis. There were 94 active preclinical vaccine candidates and 61 active development vaccine candidates. We classified the included pathogens into the following four groups: Group A consists of pathogens for which vaccines already exist-ie, Salmonella enterica serotype Typhi, Streptococcus pneumoniae, Haemophilus influenzae type b, and M tuberculosis. Group B consists of pathogens with vaccines in advanced clinical development-ie, extra-intestinal pathogenic Escherichia coli, Salmonella enterica serotype Paratyphi A, Neisseria gonorrhoeae, and C difficile. Group C consists of pathogens with vaccines in early phases of clinical development-ie, enterotoxigenic E coli, Klebsiella pneumoniae, non-typhoidal Salmonella, Shigella spp, and Campylobacter spp. Finally, group D includes pathogens with either no candidates in clinical development or low development feasibility-ie, Pseudomonas aeruginosa, Acinetobacter baumannii, Staphylococcus aureus, Helicobacter pylori, Enterococcus faecium, and Enterobacter spp. Vaccines are already important tools in reducing antimicrobial resistance and future development will provide further opportunities to optimise the use of vaccines against resistance.
Collapse
Affiliation(s)
- Isabel Frost
- World Health Organization, Geneva, Switzerland; Department of Infectious Disease, Imperial College London, London, UK.
| | - Hatim Sati
- World Health Organization, Geneva, Switzerland
| | | | | | - Christian Lienhardt
- Unité Mixte Internationale 233 IRD-U1175 INSERM, Université de Montpellier, Institut de Recherche pour le Développement, Montpellier, France; Epidemiology and Population Health, Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | | | - Peter Beyer
- World Health Organization, Geneva, Switzerland; Global Antibiotic Research and Development Partnership, Geneva, Switzerland
| |
Collapse
|