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
|
Matos ADS, Soares IF, Rodrigues-da-Silva RN, Rodolphi CM, Albrecht L, Donassolo RA, Lopez-Camacho C, Ano Bom APD, Neves PCDC, Conte FDP, Pratt-Riccio LR, Daniel-Ribeiro CT, Totino PRR, Lima-Junior JDC. Immunogenicity of PvCyRPA, PvCelTOS and Pvs25 chimeric recombinant protein of Plasmodium vivax in murine model. Front Immunol 2024; 15:1392043. [PMID: 38962015 PMCID: PMC11219565 DOI: 10.3389/fimmu.2024.1392043] [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: 02/26/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
In the Americas, P. vivax is the predominant causative species of malaria, a debilitating and economically significant disease. Due to the complexity of the malaria parasite life cycle, a vaccine formulation with multiple antigens expressed in various parasite stages may represent an effective approach. Based on this, we previously designed and constructed a chimeric recombinant protein, PvRMC-1, composed by PvCyRPA, PvCelTOS, and Pvs25 epitopes. This chimeric protein was strongly recognized by naturally acquired antibodies from exposed population in the Brazilian Amazon. However, there was no investigation about the induced immune response of PvRMC-1. Therefore, in this work, we evaluated the immunogenicity of this chimeric antigen formulated in three distinct adjuvants: Stimune, AddaVax or Aluminum hydroxide (Al(OH)3) in BALB/c mice. Our results suggested that the chimeric protein PvRMC-1 were capable to generate humoral and cellular responses across all three formulations. Antibodies recognized full-length PvRMC-1 and linear B-cell epitopes from PvCyRPA, PvCelTOS, and Pvs25 individually. Moreover, mice's splenocytes were activated, producing IFN-γ in response to PvCelTOS and PvCyRPA peptide epitopes, affirming T-cell epitopes in the antigen. While aluminum hydroxide showed notable cellular response, Stimune and Addavax induced a more comprehensive immune response, encompassing both cellular and humoral components. Thus, our findings indicate that PvRMC-1 would be a promising multistage vaccine candidate that could advance to further preclinical studies.
Collapse
MESH Headings
- Animals
- Plasmodium vivax/immunology
- Plasmodium vivax/genetics
- Mice
- Antigens, Protozoan/immunology
- Antigens, Protozoan/genetics
- Malaria, Vivax/immunology
- Malaria, Vivax/prevention & control
- Antibodies, Protozoan/immunology
- Mice, Inbred BALB C
- Malaria Vaccines/immunology
- Female
- Protozoan Proteins/immunology
- Protozoan Proteins/genetics
- Epitopes, B-Lymphocyte/immunology
- Epitopes, B-Lymphocyte/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/genetics
- Disease Models, Animal
- Adjuvants, Immunologic
- Immunogenicity, Vaccine
- Antigens, Surface
Collapse
Affiliation(s)
- Ada da Silva Matos
- Immunoparasitology Laboratory, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Isabela Ferreira Soares
- Immunoparasitology Laboratory, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | | | | | - Letusa Albrecht
- Apicomplexa Research Laboratory, Carlos Chagas Institute, Curitiba, Brazil
| | | | - Cesar Lopez-Camacho
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Ana Paula Dinis Ano Bom
- Immunological Technology Laboratory, Immunobiological Technology Institute (Bio-Manguinhos/Fiocruz), Rio de Janeiro, Brazil
| | | | - Fernando de Paiva Conte
- Eukaryotic Pilot Laboratory, Immunobiological Technology Institute (Bio-Manguinhos/Fiocruz), Rio de Janeiro, Brazil
| | | | | | | | - Josué da Costa Lima-Junior
- Immunoparasitology Laboratory, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| |
Collapse
|
3
|
Miura K. How to Accelerate Early Stage of Malaria Vaccine Development by Optimizing Functional Assays. Vaccines (Basel) 2024; 12:586. [PMID: 38932315 PMCID: PMC11209467 DOI: 10.3390/vaccines12060586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
While two Plasmodium falciparum circumsporozoite protein-based pre-erythrocytic vaccines (PEV), RTS,S and R21, have been approved by the WHO, no blood-stage vaccine (BSV) or transmission-blocking vaccine (TBV) has reached a phase 3 trial. One of the major obstacles that slows down malaria vaccine development is the shortage (or lack) of in vitro assays or animal models by which investigators can reasonably select the best vaccine formulation (e.g., antigen, adjuvant, or platform) and/or immunization strategy (e.g., interval of inoculation or route of immunization) before a human phase 2 trial. In the case of PEV, RTS,S and R21 have set a benchmark, and a new vaccine can be compared with (one of) the approved PEV directly in preclinical or early clinical studies. However, such an approach cannot be utilized for BSV or TBV development at this moment. The focus of this review is in vitro assays or in vivo models that can be used for P. falciparum BSV or TBV development, and I discuss important considerations during assay selection, standardization, qualification, validation, and interpretation of the assay results. Establishment of a robust assay/model with proper interpretation of the results is the one of key elements to accelerate future vaccine development.
Collapse
Affiliation(s)
- Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
4
|
Kuamsab N, Putaporntip C, Kakino A, Kosuwin R, Songsaigath S, Tachibana H, Jongwutiwes S. Anti-Plasmodium vivax merozoite surface protein 3 ϒ (PvMSP3 ϒ) antibodies upon natural infection. Sci Rep 2024; 14:9595. [PMID: 38671033 PMCID: PMC11053162 DOI: 10.1038/s41598-024-59153-w] [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: 01/13/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Merozoite surface protein 3 of Plasmodium vivax (PvMSP3) contains a repertoire of protein members with unique sequence organization. While the biological functions of these proteins await elucidation, PvMSP3 has been suggested to be potential vaccine targets. To date, studies on natural immune responses to this protein family have been confined to two members, PvMSP3α and PvMSP3β. This study analyzed natural IgG antibody responses to PvMSP3γ recombinant proteins derived from two variants: one containing insert blocks (CT1230nF) and the other without insert domain (NR25nF). The former variant was also expressed as two subfragment proteins: one encompassing variable domain I and insert block A (CT1230N) and the other spanning from insert block B to conserved block III (CT1230C). Serum samples were obtained from 246 symptomatic vivax malaria patients in Tak (n = 50) and Ubon Ratchathani (n = 196) Provinces. In total, 176 (71.5%) patients could mount antibodies to at least one recombinant PvMSP3γ antigen. IgG antibodies directed against antigens CT1230nF, CT1230N, CT1230C and NR25nF occurred in 96.6%, 61.4%, 71.6% and 68.2% of samples, respectively, suggesting the widespread occurrence of B-cell epitopes across PvMSP3γ. The rates of seropositivity seemed to correlate with the number of previous malaria episodes. Isotype analysis of anti-PvMSP3γ antibodies has shown predominant cytophilic subclass responses, accounting for 75.4-81.7% for IgG1 and 63.6-77.5% for IgG3. Comparing with previous studies in the same cohort, the numbers of serum samples reactive to antigens derived from P. vivax merozoite surface protein 9 (PvMSP9) and thrombospondin-related anonymous protein (PvTRAP) were higher than those to PvMSP3γ, being 92.7% and 87.0% versus 71.5%, respectively. Three (1.22%) serum samples were nonresponsive to all these malarial proteins. Nevertheless, the relevance of naturally acquired antibodies to PvMSP3γ in host protection requires further studies.
Collapse
Affiliation(s)
- Napaporn Kuamsab
- Molecular Biology of Malaria and Opportunistic Parasites Research Unit, Department of Parasitology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Infectious Diseases, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Community Public Health Program, Faculty of Health Science and Technology, Southern College of Technology, Nakorn Si Thammarat, Thailand
| | - Chaturong Putaporntip
- Molecular Biology of Malaria and Opportunistic Parasites Research Unit, Department of Parasitology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| | - Azumi Kakino
- Department of Infectious Diseases, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Rattiporn Kosuwin
- Molecular Biology of Malaria and Opportunistic Parasites Research Unit, Department of Parasitology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Infectious Diseases, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Department of Health Promotion, Faculty of Physical Therapy, Srinakharinwirot University, Nakhonnayok, Thailand
| | - Sunisa Songsaigath
- Molecular Biology of Malaria and Opportunistic Parasites Research Unit, Department of Parasitology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Infectious Diseases, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Department of Health Promotion, Faculty of Physical Therapy, Srinakharinwirot University, Nakhonnayok, Thailand
| | - Hiroshi Tachibana
- Department of Infectious Diseases, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Somchai Jongwutiwes
- Molecular Biology of Malaria and Opportunistic Parasites Research Unit, Department of Parasitology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
5
|
Tiono AB, Plieskatt JL, Ouedraogo A, Soulama BI, Miura K, Bougouma EC, Naghizadeh M, Barry A, Yaro JBB, Ezinmegnon S, Henry N, Ofori EA, Adu B, Singh SK, Konkobo A, Lövgren Bengtsson K, Diarra A, Carnrot C, Reimer JM, Ouedraogo A, Tienta M, Long CA, Ouedraogo IN, Sagara I, Sirima SB, Theisen M. A randomized first-in-human phase I trial of differentially adjuvanted Pfs48/45 malaria vaccines in Burkinabé adults. J Clin Invest 2024; 134:e175707. [PMID: 38290009 PMCID: PMC10977980 DOI: 10.1172/jci175707] [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: 09/11/2023] [Accepted: 01/26/2024] [Indexed: 02/01/2024] Open
Abstract
BACKGROUNDMalaria transmission-blocking vaccines aim to interrupt the transmission of malaria from one person to another.METHODSThe candidates R0.6C and ProC6C share the 6C domain of the Plasmodium falciparum sexual-stage antigen Pfs48/45. R0.6C utilizes the glutamate-rich protein (GLURP) as a carrier, and ProC6C includes a second domain (Pfs230-Pro) and a short 36-amino acid circumsporozoite protein (CSP) sequence. Healthy adults (n = 125) from a malaria-endemic area of Burkina Faso were immunized with 3 intramuscular injections, 4 weeks apart, of 30 μg or 100 μg R0.6C or ProC6C each adsorbed to Alhydrogel (AlOH) adjuvant alone or in combination with Matrix-M (15 μg or 50 μg, respectively). The allocation was random and double-blind for this phase I trial.RESULTSThe vaccines were safe and well tolerated with no vaccine-related serious adverse events. A total of 7 adverse events, mild to moderate in intensity and considered possibly related to the study vaccines, were recorded. Vaccine-specific antibodies were highest in volunteers immunized with 100 μg ProC6C-AlOH with Matrix-M, and 13 of 20 (65%) individuals in the group showed greater than 80% transmission-reducing activity (TRA) when evaluated in the standard membrane feeding assay at 15 mg/mL IgG. In contrast, R0.6C induced sporadic TRA.CONCLUSIONAll formulations were safe and well tolerated in a malaria-endemic area of Africa in healthy adults. The ProC6C-AlOH/Matrix-M vaccine elicited the highest levels of functional antibodies, meriting further investigation.TRIAL REGISTRATIONPan-African Clinical Trials Registry (https://pactr.samrc.ac.za) PACTR202201848463189.FUNDINGThe study was funded by the European and Developing Countries Clinical Trials Partnership (grant RIA2018SV-2311).
Collapse
Affiliation(s)
- Alfred B. Tiono
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Jordan L. Plieskatt
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
| | | | | | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Edith C. Bougouma
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Mohammad Naghizadeh
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Aissata Barry
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | | | - Sem Ezinmegnon
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Noelie Henry
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Ebenezer Addo Ofori
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Bright Adu
- Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Susheel K. Singh
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Augustin Konkobo
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | | | - Amidou Diarra
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | | | | | - Amidou Ouedraogo
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Moussa Tienta
- Malaria Research and Training Center, Mali–National Institute of Allergy and Infectious Diseases International Center for Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Carole A. Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Issa N. Ouedraogo
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Issaka Sagara
- Malaria Research and Training Center, Mali–National Institute of Allergy and Infectious Diseases International Center for Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | | | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Palacpac NMQ, Ishii KJ, Arisue N, Tougan T, Horii T. Immune tolerance caused by repeated P. falciparum infection against SE36 malaria vaccine candidate antigen and the resulting limited polymorphism. Parasitol Int 2024; 99:102845. [PMID: 38101534 DOI: 10.1016/j.parint.2023.102845] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/27/2023] [Accepted: 12/10/2023] [Indexed: 12/17/2023]
Abstract
The call for second generation malaria vaccines needs not only the identification of novel candidate antigens or adjuvants but also a better understanding of immune responses and the underlying protective processes. Plasmodium parasites have evolved a range of strategies to manipulate the host immune system to guarantee survival and establish parasitism. These immune evasion strategies hamper efforts to develop effective malaria vaccines. In the case of a malaria vaccine targeting the N-terminal domain of P. falciparum serine repeat antigen 5 (SE36), now in clinical trials, we observed reduced responsiveness (lowered immunogenicity) which may be attributed to immune tolerance/immune suppression. Here, immunogenicity data and insights into the immune responses to SE36 antigen from epidemiological studies and clinical trials are summarized. Documenting these observations is important to help identify gaps for SE36 continued development and engender hope that highly effective blood-stage/multi-stage vaccines can be achieved.
Collapse
Affiliation(s)
- Nirianne Marie Q Palacpac
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.
| | - Ken J Ishii
- Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan; Laboratory of Vaccine Science, Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan; Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan.
| | - Nobuko Arisue
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.
| | - Takahiro Tougan
- Department of Molecular Protozoology, 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
|
7
|
Plieskatt J, Bang P, Wood GK, Naghizadeh M, Singh SK, Jore MM, Theisen M. Clinical formulation development of Plasmodium falciparum malaria vaccine candidates based on Pfs48/45, Pfs230, and PfCSP. Vaccine 2024; 42:1980-1992. [PMID: 38388238 DOI: 10.1016/j.vaccine.2024.02.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/09/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024]
Abstract
Two malaria transmission-blocking vaccine (TBV) candidates, R0.6C and ProC6C, have completed preclinical development including the selection of adjuvants, Alhydrogel® with or without the saponin based adjuvant Matrix-M™. Here, we report on the final drug product (formulation) design of R0.6C and ProC6C and evaluate their safety and biochemical stability in preparation for preclinical and clinical pharmacy handling. The point-of-injection stability studies demonstrated that both the R0.6C and ProC6C antigens are stable on Alhydrogel in the presence or absence of Matrix-M for up to 24 h at room temperature. As this is the first study to combine Alhydrogel and Matrix-M for clinical use, we also evaluated their potential interactions. Matrix-M adsorbs to Alhydrogel, while not displacing the > 95 % adsorbed protein. The R0.6C and ProC6C formulations were found to be safe and well tolerated in repeated dose toxicity studies in rabbits generating high levels of functional antibodies that blocked infection of mosquitoes. Further, the R0.6C and ProC6C drug products were found to be stable for minimally 24 months when stored at 2-8 °C, with studies ongoing through 36 months. Together, this data demonstrates the safety and suitability of the L. lactis expression system as well as supports the clinical testing of the R0.6C and ProC6C malaria vaccine candidates in First-In-Human clinical trials.
Collapse
Affiliation(s)
- Jordan Plieskatt
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Bang
- Department of Vaccine Development, Statens Serum Institut, Copenhagen, Denmark
| | - Grith Krøyer Wood
- Department of Vaccine Development, Statens Serum Institut, Copenhagen, Denmark
| | - Mohammad Naghizadeh
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Susheel K Singh
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Matthijs M Jore
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
8
|
Takashima E, Otsuki H, Morita M, Ito D, Nagaoka H, Yuguchi T, Hassan I, Tsuboi T. The Need for Novel Asexual Blood-Stage Malaria Vaccine Candidates for Plasmodium falciparum. Biomolecules 2024; 14:100. [PMID: 38254700 PMCID: PMC10813614 DOI: 10.3390/biom14010100] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/25/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Extensive control efforts have significantly reduced malaria cases and deaths over the past two decades, but in recent years, coupled with the COVID-19 pandemic, success has stalled. The WHO has urged the implementation of a number of interventions, including vaccines. The modestly effective RTS,S/AS01 pre-erythrocytic vaccine has been recommended by the WHO for use in sub-Saharan Africa against Plasmodium falciparum in children residing in moderate to high malaria transmission regions. A second pre-erythrocytic vaccine, R21/Matrix-M, was also recommended by the WHO on 3 October 2023. However, the paucity and limitations of pre-erythrocytic vaccines highlight the need for asexual blood-stage malaria vaccines that prevent disease caused by blood-stage parasites. Few asexual blood-stage vaccine candidates have reached phase 2 clinical development, and the challenges in terms of their efficacy include antigen polymorphisms and low immunogenicity in humans. This review summarizes the history and progress of asexual blood-stage malaria vaccine development, highlighting the need for novel candidate vaccine antigens/molecules.
Collapse
Affiliation(s)
- Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama 790-8577, Japan; (M.M.); (H.N.); (T.Y.); (I.H.)
| | - Hitoshi Otsuki
- Division of Medical Zoology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan; (H.O.); (D.I.)
| | - Masayuki Morita
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama 790-8577, Japan; (M.M.); (H.N.); (T.Y.); (I.H.)
| | - Daisuke Ito
- Division of Medical Zoology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan; (H.O.); (D.I.)
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama 790-8577, Japan; (M.M.); (H.N.); (T.Y.); (I.H.)
| | - Takaaki Yuguchi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama 790-8577, Japan; (M.M.); (H.N.); (T.Y.); (I.H.)
| | - Ifra Hassan
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama 790-8577, Japan; (M.M.); (H.N.); (T.Y.); (I.H.)
| | - Takafumi Tsuboi
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama 790-8577, Japan
| |
Collapse
|
9
|
Rajneesh, Tiwari R, Singh VK, Kumar A, Gupta RP, Singh AK, Gautam V, Kumar R. Advancements and Challenges in Developing Malaria Vaccines: Targeting Multiple Stages of the Parasite Life Cycle. ACS Infect Dis 2023; 9:1795-1814. [PMID: 37708228 DOI: 10.1021/acsinfecdis.3c00332] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Malaria, caused by Plasmodium species, remains a major global health concern, causing millions of deaths annually. While the introduction of the RTS,S vaccine has shown promise, there is a pressing need for more effective vaccines due to the emergence of drug-resistant parasites and insecticide-resistant vectors. However, the complex life cycle and genetic diversity of the parasite, technical obstacles, limited funding, and the impact of the 2019 pandemic have hindered progress in malaria vaccine development. This review focuses on advancements in malaria vaccine development, particularly the ongoing clinical trials targeting antigens from different stages of the Plasmodium life cycle. Additionally, we discuss the rationale, strategies, and challenges associated with vaccine design, aiming to enhance the immune response and protective efficacy of vaccine candidates. A cost-effective and multistage vaccine could hold the key to controlling and eradicating malaria.
Collapse
Affiliation(s)
- Rajneesh
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rahul Tiwari
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Vishal K Singh
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Awnish Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rohit P Gupta
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
- Department of Applied Microbiology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Akhilesh K Singh
- Faculty of Dental Science, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Vibhav Gautam
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rajiv Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
10
|
El-Moamly AA, El-Sweify MA. Malaria vaccines: the 60-year journey of hope and final success-lessons learned and future prospects. Trop Med Health 2023; 51:29. [PMID: 37198702 DOI: 10.1186/s41182-023-00516-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/18/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND The world has made great strides towards beating malaria, although about half of the world population is still exposed to the risk of contracting malaria. Developing an effective malaria vaccine was a huge challenge for medical science. In 2021 the World Health Organization (WHO) approved the first malaria vaccine, RTS,S/AS01 vaccine (Mosquirix™), for widespread use. This review highlights the history of development, and the different approaches and types of malaria vaccines, and the literature to date. It covers the developmental stages of RTS,S/AS01 and recommends steps for its deployment. The review explores other potential vaccine candidates and their status, and suggests options for their further development. It also recommends future roles for vaccines in eradicating malaria. Questions remain on how RTS,S vaccine will work in widespread use and how it can best be utilized to benefit vulnerable communities. CONCLUSION Malaria vaccines have been in development for almost 60 years. The RTS,S/AS01 vaccine has now been approved, but cannot be a stand-alone solution. Development should continue on promising candidates such as R21, PfSPZ and P. vivax vaccines. Multi-component vaccines may be a useful addition to other malaria control techniques in achieving eradication of malaria.
Collapse
Affiliation(s)
- Amal A El-Moamly
- Department of Medical Parasitology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Mohamed A El-Sweify
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
11
|
Singh SK, Naghizadeh M, Plieskatt J, Singh S, Theisen M. Cloning and Recombinant Protein Expression in Lactococcus lactis. Methods Mol Biol 2023; 2652:3-20. [PMID: 37093467 DOI: 10.1007/978-1-0716-3147-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
The Lactococcus lactis, a Gram-positive bacteria, is an ideal expression host for the overproduction of heterologous proteins in a properly folded and functional form. L. lactis has been identified as an efficient cell factory, generally recognized as safe (GRAS), has a long history of safe use in food production, and is known to have probiotic properties. Key desirable features of L. lactis include the following: (1) rapid growth to high cell densities, not requiring aeration which facilitates large-scale fermentation; (2) its Gram-positive nature precludes the presence of contaminating endotoxins; (3) the capacity to secrete stable recombinant protein into the growth medium with few proteases resulting in a properly folded, full-length protein; and (4) the availability of diverse expression vectors facilitating various cloning options. We have previously described production of several recombinant proteins with varying degrees of predicted structural complexities using the L. lactis pH-dependent P170 promoter. The purpose of this chapter is to provide a detailed protocol for facilitating wider application of L. lactis as a reliable platform for expression of heterologous recombinant proteins in soluble form. Here, we present details of the various steps involved such as cloning of the target gene in appropriate expression plasmid vector, determination of the expression levels of the heterologous protein, and initial purification of the expressed soluble recombinant protein of interest.
Collapse
Affiliation(s)
- Susheel K Singh
- Biotherapeutic and Vaccine Research Division, ICMR-Regional Medical Research Centre, Bhubaneswar, Odisha, India
| | - Mohammad Naghizadeh
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jordan Plieskatt
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Subhash Singh
- Biotherapeutic and Vaccine Research Division, ICMR-Regional Medical Research Centre, Bhubaneswar, Odisha, India
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
12
|
Adukpo S, Adedoja A, Esen M, Theisen M, Ntoumi F, Ojurongbe O. Humoral antimalaria immune response in Nigerian children exposed to helminth and malaria parasites. Front Immunol 2022; 13:979727. [PMID: 36159869 PMCID: PMC9494551 DOI: 10.3389/fimmu.2022.979727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/16/2022] [Indexed: 11/14/2022] Open
Abstract
Background Malaria and helminthic parasites are endemic in tropical countries, and co-infections might influence host-parasite interactions. In this community-based cross-sectional study, the effect that the presence of soil-transmitted helminths (STH) (Hookworm, Hymenolepis nana) and Schistosoma haematobium infections could have on the immunoglobulin (Ig) candidate protein of the malaria vaccine GMZ2 levels was evaluated. Methods Blood, stool, and urine samples were collected from 5-15-year-old children to diagnose P. falciparum (Pf), STH, and Schistosoma haematobium, respectively. Identification and quantification of the parasite load of STH and S. haematobium were achieved by light microscopy. A polymerase chain reaction was carried out to detect submicroscopic infections of P. falciparum. Plasma levels of GMZ2 specific IgG and its subclasses were quantified by ELISA. Results The median level of total IgG in individuals with co-infection with Pf/H. nana was significantly lower in the mono-infected group with Pf (p = 0.0121) or study participants without infection (p=0.0217). Similarly, the median level of IgG1 was statistically lower in Pf/H. nana group compared to Pf-group (p=0.0137). Equally, the Pf/H. nana infected individuals posted a lower level of IgG1 compared to Pf-group (p=0.0137) and IgG4 compared to the Pf-group (p=0.0144). Spearman rank correlation analyses indicated positive relationships between the densities of H. nana (ρ=0.25, p=0.015) and S. haematobium (ρ=0.36, p<0.0001). Conclusions Hookworm and H. nana infections are associated with reduced GMZ2 specific IgG levels. This study shows the possible manipulation of immune responses by helminths for their survival and transmission, which may have serious implications for vaccine development and deployment in helminth-endemic regions.
Collapse
Affiliation(s)
- Selorme Adukpo
- Department of Pharmaceutics and Microbiology, School of Pharmacy, University of Ghana, Accra, Ghana
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Ayodele Adedoja
- Department of Medical Microbiology and Parasitology, Ladoke Akintola University of Technology, Osogbo, Nigeria
- Department of Medical Microbiology and Parasitology, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Meral Esen
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Centre for Medical Parasitology at the Department of International Health, Immunology, and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Francine Ntoumi
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Infectious Disease Department, Fondation Congolaise pour la Recherche Médicale, Brazzaville, Republic of Congo
| | - Olusola Ojurongbe
- Department of Medical Microbiology and Parasitology, Ladoke Akintola University of Technology, Osogbo, Nigeria
- Centre for Emerging and Re-emerging Infectious Disease, Humboldt-Bayer Foundations Research Hub, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
- *Correspondence: Olusola Ojurongbe,
| |
Collapse
|
13
|
Dassah S, Adu B, Tiendrebeogo RW, Singh SK, Arthur FKN, Sirima SB, Theisen M. GMZ2 Vaccine-Induced Antibody Responses, Naturally Acquired Immunity and the Incidence of Malaria in Burkinabe Children. Front Immunol 2022; 13:899223. [PMID: 35720297 PMCID: PMC9200992 DOI: 10.3389/fimmu.2022.899223] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
GMZ2 is a malaria vaccine candidate evaluated in a phase 2b multi-centre trial. Here we assessed antibody responses and the association of naturally acquired immunity with incidence of malaria in one of the trial sites, Banfora in Burkina Faso. The analysis included 453 (GMZ2 = 230, rabies = 223) children aged 12-60 months old. Children were followed-up for clinical malaria episodes for 12 months after final vaccine administration. Antibody levels against GMZ2 and eleven non-GMZ2 antigens were measured on days 0 and 84 (one month after final vaccine dose). Vaccine efficacy (VE) differed by age group (interaction, (12-35 months compared to 36-60 months), p = 0.0615). During the twelve months of follow-up, VE was 1% (95% confidence interval [CI] -17%, 17%) and 23% ([CI] 3%, 40%) in the 12 - 35 and 36 - 60 months old children, respectively. In the GMZ2 group, day 84 anti-GMZ2 IgG levels were associated with reduced incidence of febrile malaria during the follow up periods of 1-6 months (hazard ratio (HR) = 0.87, 95%CI = (0.77, 0.98)) and 7-12 months (HR = 0.84, 95%CI = (0.71, 0.98)) in the 36-60 months old but not in 12-35 months old children. Multivariate analysis involving day 84 IgG levels to eleven non-vaccine antigens, identified MSP3-K1 and GLURP-R2 to be associated with reduced incidence of malaria during the 12 months of follow up. The inclusion of these antigens might improve GMZ2 vaccine efficacy.
Collapse
Affiliation(s)
- Sylvester Dassah
- Navrongo Health Research Centre, Navrongo, Ghana.,Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Bright Adu
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Régis W Tiendrebeogo
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of Immunology, and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Susheel K Singh
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of Immunology, and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Fareed K N Arthur
- Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Sodiomon B Sirima
- Groupe de Recherche Action en Senté (GRAS), Ouagadougou, Burkina Faso
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of Immunology, and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Baptista BO, de Souza ABL, Riccio EKP, Bianco-Junior C, Totino PRR, Martins da Silva JH, Theisen M, Singh SK, Amoah LE, Ribeiro-Alves M, Souza RM, Lima-Junior JC, Daniel-Ribeiro CT, Pratt-Riccio LR. Naturally acquired antibody response to a Plasmodium falciparum chimeric vaccine candidate GMZ2.6c and its components (MSP-3, GLURP, and Pfs48/45) in individuals living in Brazilian malaria-endemic areas. Malar J 2022; 21:6. [PMID: 34983540 PMCID: PMC8729018 DOI: 10.1186/s12936-021-04020-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 12/09/2021] [Indexed: 12/19/2022] Open
Abstract
Background The GMZ2.6c malaria vaccine candidate is a multi-stage Plasmodium falciparum chimeric protein which contains a fragment of the sexual-stage Pfs48/45-6C protein genetically fused to GMZ2, a fusion protein of GLURP and MSP-3, that has been shown to be well tolerated, safe and immunogenic in clinical trials performed in a malaria-endemic area of Africa. However, there is no data available on the antigenicity or immunogenicity of GMZ2.6c in humans. Considering that circulating parasites can be genetically distinct in different malaria-endemic areas and that host genetic factors can influence the immune response to vaccine antigens, it is important to verify the antigenicity, immunogenicity and the possibility of associated protection in individuals living in malaria-endemic areas with different epidemiological scenarios. Herein, the profile of antibody response against GMZ2.6c and its components (MSP-3, GLURP and Pfs48/45) in residents of the Brazilian Amazon naturally exposed to malaria, in areas with different levels of transmission, was evaluated. Methods This study was performed using serum samples from 352 individuals from Cruzeiro do Sul and Mâncio Lima, in the state of Acre, and Guajará, in the state of Amazonas. Specific IgG, IgM, IgA and IgE antibodies and IgG subclasses were detected by Enzyme-Linked Immunosorbent Assay. Results The results showed that GMZ2.6c protein was widely recognized by naturally acquired antibodies from individuals of the Brazilian endemic areas with different levels of transmission. The higher prevalence of individuals with antibodies against GMZ2.6c when compared to its individual components may suggest an additive effect of GLURP, MSP-3, and Pfs48/45 when inserted in a same construct. Furthermore, naturally malaria-exposed individuals predominantly had IgG1 and IgG3 cytophilic anti-GMZ2.6c antibodies, an important fact considering that the acquisition of anti-malaria protective immunity results from a delicate balance between cytophilic/non-cytophilic antibodies. Interestingly, anti-GMZ2.6c antibodies seem to increase with exposure to malaria infection and may contribute to parasite immunity. Conclusions The data showed that GMZ2.6c protein is widely recognized by naturally acquired antibodies from individuals living in malaria-endemic areas in Brazil and that these may contribute to parasite immunity. These data highlight the importance of GMZ2.6c as a candidate for an anti-malarial vaccine. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-021-04020-6.
Collapse
Affiliation(s)
- Barbara Oliveira Baptista
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil.,Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brazil
| | - Ana Beatriz Lopes de Souza
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil.,Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brazil
| | - Evelyn Kety Pratt Riccio
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil.,Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brazil
| | - Cesare Bianco-Junior
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil.,Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brazil
| | - Paulo Renato Rivas Totino
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil.,Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brazil
| | | | - Michael Theisen
- Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Susheel Kumar Singh
- Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Linda Eva Amoah
- Immunology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Marcelo Ribeiro-Alves
- Laboratório de Pesquisa Clínica em DST e AIDS, Instituto Nacional de Infectologia Evandro Chagas, Fiocruz, Rio de Janeiro, Brazil
| | - Rodrigo Medeiros Souza
- Laboratório de Doenças Infecciosas na Amazônia Ocidental, Universidade Federal do Acre, Acre, Brazil
| | | | - Cláudio Tadeu Daniel-Ribeiro
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil.,Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brazil
| | - Lilian Rose Pratt-Riccio
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil. .,Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brazil.
| |
Collapse
|
15
|
Pirahmadi S, Afzali S, Zargar M, Zakeri S, Mehrizi AA. How can we develop an effective subunit vaccine to achieve successful malaria eradication? Microb Pathog 2021; 160:105203. [PMID: 34547408 DOI: 10.1016/j.micpath.2021.105203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/05/2021] [Accepted: 09/17/2021] [Indexed: 12/16/2022]
Abstract
Malaria, a mosquito-borne infection, is the most widespread parasitic disease. Despite numerous efforts to eradicate malaria, this disease is still a health concern worldwide. Owing to insecticide-resistant vectors and drug-resistant parasites, available controlling measures are insufficient to achieve a malaria-free world. Thus, there is an urgent need for new intervention tools such as efficient malaria vaccines. Subunit vaccines are the most promising malaria vaccines under development. However, one of the major drawbacks of subunit vaccines is the lack of efficient and durable immune responses including antigen-specific antibody, CD4+, and CD8+ T-cell responses, long-lived plasma cells, memory cells, and functional antibodies for parasite neutralization or inhibition of parasite invasion. These types of responses could be induced by whole organism vaccines, but eliciting these responses with subunit vaccines has been proven to be more challenging. Consequently, subunit vaccines require several policies to overcome these challenges. In this review, we address common approaches that can improve the efficacy of subunit vaccines against malaria.
Collapse
Affiliation(s)
- Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Shima Afzali
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Mostafa Zargar
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Akram Abouie Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
16
|
Opi DH, Kurtovic L, Chan JA, Horton JL, Feng G, Beeson JG. Multi-functional antibody profiling for malaria vaccine development and evaluation. Expert Rev Vaccines 2021; 20:1257-1272. [PMID: 34530671 DOI: 10.1080/14760584.2021.1981864] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION A vaccine would greatly accelerate current global efforts toward malaria elimination. While a partially efficacious vaccine has been achieved for Plasmodium falciparum, a major bottleneck in developing highly efficacious vaccines is a lack of reliable correlates of protection, and the limited application of assays that quantify functional immune responses to evaluate and down-select vaccine candidates in pre-clinical studies and clinical trials. AREAS COVERED In this review, we describe the important role of antibodies in immunity against malaria and detail the nature and functional activities of antibodies against the malaria-causing parasite. We highlight the growing understanding of antibody effector functions against malaria and in vitro assays to measure these functional antibody responses. We discuss the application of these assays to quantify antibody functions in vaccine development and evaluation. EXPERT OPINION It is becoming increasingly clear that multiple antibody effector functions are involved in immunity to malaria. Therefore, we propose that evaluating vaccine candidates needs to move beyond individual assays or measuring IgG magnitude alone. Instead, vaccine evaluation should incorporate the full breadth of antibody response types and harness a wider range of assays measuring functional antibody responses. We propose a 3-tier approach to implementing assays to inform vaccine evaluation.
Collapse
Affiliation(s)
- D Herbert Opi
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Liriye Kurtovic
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia
| | - Jo-Anne Chan
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Jessica L Horton
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Gaoqian Feng
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - James G Beeson
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia.,Department of Microbiology, Monash University, Clayton, Australia
| |
Collapse
|
17
|
Abstract
Introduction: An effective vaccine against malaria forms a global health priority. Both naturally acquired immunity and sterile protection induced by irradiated sporozoite immunization were described decades ago. Still no vaccine exists that sufficiently protects children in endemic areas. Identifying immunological correlates of vaccine efficacy can inform rational vaccine design and potentially accelerate clinical development.Areas covered: We discuss recent research on immunological correlates of malaria vaccine efficacy, including: insights from state-of-the-art omics platforms and systems vaccinology analyses; functional anti-parasitic assays; pre-immunization predictors of vaccine efficacy; and comparison of correlates of vaccine efficacy against controlled human malaria infections (CHMI) and against naturally acquired infections.Expert Opinion: Effective vaccination may be achievable without necessarily understanding immunological correlates, but the relatively disappointing efficacy of malaria vaccine candidates in target populations is concerning. Hypothesis-generating omics and systems vaccinology analyses, alongside assessment of pre-immunization correlates, have the potential to bring about paradigm-shifts in malaria vaccinology. Functional assays may represent in vivo effector mechanisms, but have scarcely been formally assessed as correlates. Crucially, evidence is still meager that correlates of vaccine efficacy against CHMI correspond with those against naturally acquired infections in target populations. Finally, the diversity of immunological assays and efficacy endpoints across malaria vaccine trials remains a major confounder.
Collapse
Affiliation(s)
| | - Matthew B B McCall
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, The Netherlands.,Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| |
Collapse
|
18
|
Hamre KES, Ondigo BN, Hodges JS, Dutta S, Theisen M, Ayodo G, John CC. Antibody Correlates of Protection from Clinical Plasmodium falciparum Malaria in an Area of Low and Unstable Malaria Transmission. Am J Trop Med Hyg 2020; 103:2174-2182. [PMID: 33124533 DOI: 10.4269/ajtmh.18-0805] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Immune correlates of protection against clinical malaria are difficult to ascertain in low-transmission areas because of the limited number of malaria cases. We collected blood samples from 5,753 individuals in a Kenyan highland area, ascertained malaria incidence in this population over the next 6 years, and then compared antibody responses to 11 Plasmodium falciparum vaccine candidate antigens in individuals who did versus did not develop clinical malaria in a nested case-control study (154 cases and 462 controls). Individuals were matched by age and village. Antigens tested included circumsporozoite protein (CSP), liver-stage antigen (LSA)-1, apical membrane antigen-1 FVO and 3D7 strains, erythrocyte-binding antigen-175, erythrocyte-binding protein-2, merozoite surface protein (MSP)-1 FVO and 3D7 strains, MSP-3, and glutamate-rich protein (GLURP) N-terminal non-repetitive (R0) and C-terminal repetitive (R2) regions. After adjustment for potential confounding factors, the presence of antibodies to LSA-1, GLURP-R2, or GLURP-R0 was associated with decreased odds of developing clinical malaria (odds ratio [OR], [95% CI] 0.56 [0.36-0.89], 0.56 [0.36-0.87], and 0.77 [0.43-1.02], respectively). Levels of antibodies to LSA-1, GLURP-R2, and CSP were associated with decreased odds of developing clinical malaria (OR [95% CI]; 0.61 [0.41-0.89], 0.60 [0.43-0.84], and 0.49 [0.24-0.99], for every 10-fold increase in antibody levels, respectively). The presence of antibodies to CSP, GLURP-R0, GLURP-R2, and LSA-1 combined best-predicted protection from clinical malaria. Antibodies to CSP, GLURP-R0, GLURP-R2, and LSA-1 are associated with protection against clinical malaria in a low-transmission setting. Vaccines containing these antigens should be evaluated in low malaria transmission areas.
Collapse
Affiliation(s)
- Karen E S Hamre
- CDC Foundation, Atlanta, Georgia.,Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota.,Division of Global Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Bartholomew N Ondigo
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland.,Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya.,Department of Biochemistry and Molecular Biology, Egerton University, Nakuru, Kenya
| | - James S Hodges
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota
| | - Sheetij Dutta
- Walter Reed Army Institute for Research, Silver Spring, Maryland
| | | | - George Ayodo
- Jaramogi Oginga Odinga University of Science and Technology, Bondo, Kenya.,Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Chandy C John
- Division of Global Pediatrics, University of Minnesota, Minneapolis, Minnesota.,Department of Biochemistry and Molecular Biology, Egerton University, Nakuru, Kenya.,Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota.,Department of Pediatrics, Indiana University, Indianapolis, Indiana
| |
Collapse
|
19
|
Skwarczynski M, Chandrudu S, Rigau-Planella B, Islam MT, Cheong YS, Liu G, Wang X, Toth I, Hussein WM. Progress in the Development of Subunit Vaccines against Malaria. Vaccines (Basel) 2020; 8:vaccines8030373. [PMID: 32664421 PMCID: PMC7563759 DOI: 10.3390/vaccines8030373] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/02/2022] Open
Abstract
Malaria is a life-threatening disease and one of the main causes of morbidity and mortality in the human population. The disease also results in a major socio-economic burden. The rapid spread of malaria epidemics in developing countries is exacerbated by the rise in drug-resistant parasites and insecticide-resistant mosquitoes. At present, malaria research is focused mainly on the development of drugs with increased therapeutic effects against Plasmodium parasites. However, a vaccine against the disease is preferable over treatment to achieve long-term control. Trials to develop a safe and effective immunization protocol for the control of malaria have been occurring for decades, and continue on today; still, no effective vaccines are available on the market. Recently, peptide-based vaccines have become an attractive alternative approach. These vaccines utilize short protein fragments to induce immune responses against malaria parasites. Peptide-based vaccines are safer than traditional vaccines, relatively inexpensive to produce, and can be composed of multiple T- and B-cell epitopes integrated into one antigenic formulation. Various combinations, based on antigen choice, peptide epitope modification and delivery mechanism, have resulted in numerous potential malaria vaccines candidates; these are presently being studied in both preclinical and clinical trials. This review describes the current landscape of peptide-based vaccines, and addresses obstacles and opportunities in the production of malaria vaccines.
Collapse
Affiliation(s)
- Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Saranya Chandrudu
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Berta Rigau-Planella
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Md. Tanjir Islam
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Yee S. Cheong
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Genan Liu
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Xiumin Wang
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4072, Australia
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- Correspondence: (I.T.); (W.M.H.)
| | - Waleed M. Hussein
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
- Correspondence: (I.T.); (W.M.H.)
| |
Collapse
|
20
|
Nouatin O, Ateba Ngoa U, Ibáñez J, Dejon-Agobe JC, Mordmüller B, Edoa JR, Mougeni F, Brückner S, Bouyoukou Hounkpatin A, Esen M, Theisen M, Moutairou K, Hoffman SL, Issifou S, Luty AJF, Loembe MM, Agnandji ST, Lell B, Kremsner PG, Adegnika AA. Effect of immune regulatory pathways after immunization with GMZ2 malaria vaccine candidate in healthy lifelong malaria-exposed adults. Vaccine 2020; 38:4263-4272. [PMID: 32386747 PMCID: PMC7297038 DOI: 10.1016/j.vaccine.2020.04.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/08/2020] [Accepted: 04/20/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Despite appreciable immunogenicity in malaria-naive populations, many candidate malaria vaccines are considerably less immunogenic in malaria-exposed populations. This could reflect induction of immune regulatory mechanisms involving Human Leukocyte Antigen G (HLA-G), regulatory T (Treg), and regulatory B (Breg) cells. Here, we addressed the question whether there is correlation between these immune regulatory pathways and both plasmablast frequencies and vaccine-specific IgG concentrations. METHODS Fifty Gabonese adults with lifelong exposure to Plasmodium spp were randomized to receive three doses of either 30 µg or 100 µg GMZ2-CAF01, or 100 µg GMZ2-alum, or control vaccine (rabies vaccine) at 4-week intervals. Only plasma and peripheral blood mononuclear cells isolated from blood samples collected before (D0) and 28 days after the third vaccination (D84) of 35 participants were used to measure sHLA-G levels and anti-GMZ2 IgG concentrations, and to quantify Treg, Breg and plasmablast cells. Vaccine efficacy was assessed using controlled human malaria infection (CHMI) by direct venous inoculation of Plasmodium falciparum sporozoites (PfSPZ Challenge). RESULTS The sHLA-G concentration increased from D0 to D84 in all GMZ2 vaccinated participants and in the control group, whereas Treg frequencies increased only in those receiving 30 µg or 100 µg GMZ2-CAF01. The sHLA-G level on D84 was associated with a decrease of the anti-GMZ2 IgG concentration, whereas Treg frequencies on D0 or on D84, and Breg frequency on D84 were associated with lower plasmablast frequencies. Importantly, having a D84:D0 ratio of sHLA-G above the median was associated with an increased risk of P. falciparum infection after sporozoites injection. CONCLUSION Regulatory immune responses are induced following immunization. Stronger sHLA-G and Treg immune responses may suppress vaccine induced immune responses, and the magnitude of the sHLA-G response increased the risk of Plasmodium falciparum infection after CHMI. These findings could have implications for the design and testing of malaria vaccine candidates in semi-immune individuals.
Collapse
Affiliation(s)
- Odilon Nouatin
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany; Germany; German Centre for Infection Research (DZIF), Partner Site Tübingen, Germany; Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d'Abomey-Calavi, Cotonou, Benin.
| | - Ulysse Ateba Ngoa
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon.
| | - Javier Ibáñez
- Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany
| | - Jean Claude Dejon-Agobe
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany
| | - Benjamin Mordmüller
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany; Germany; German Centre for Infection Research (DZIF), Partner Site Tübingen, Germany.
| | - Jean Ronald Edoa
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany.
| | - Fabrice Mougeni
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon
| | - Sina Brückner
- Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany.
| | - Aurore Bouyoukou Hounkpatin
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany
| | - Meral Esen
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany; Germany; German Centre for Infection Research (DZIF), Partner Site Tübingen, Germany.
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark and Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen, and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark.
| | - Kabirou Moutairou
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d'Abomey-Calavi, Cotonou, Benin.
| | | | - Saadou Issifou
- Fondation pour la Recherche Scientifique, 72 BP45 Cotonou, Benin.
| | - Adrian J F Luty
- Centre d'Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l'Enfance, Faculté des Sciences de la Santé, Université d'Abomey-Calavi, Cotonou, MERIT UMR D216, Benin; Université de Paris, MERIT, IRD, Paris, France.
| | - Marguerite M Loembe
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany; Germany; German Centre for Infection Research (DZIF), Partner Site Tübingen, Germany.
| | - Selidji Todagbé Agnandji
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany; Germany; German Centre for Infection Research (DZIF), Partner Site Tübingen, Germany.
| | - Bertrand Lell
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria.
| | - Peter G Kremsner
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany; Germany; German Centre for Infection Research (DZIF), Partner Site Tübingen, Germany.
| | - Ayôla Akim Adegnika
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany; Germany; German Centre for Infection Research (DZIF), Partner Site Tübingen, Germany; Leiden University Medical Centre (LUMC), 2333 ZA Leiden, the Netherlands.
| |
Collapse
|
21
|
Lê HG, Thái TL, Kang JM, Lee J, Moe M, Võ TC, Naw H, Myint MK, Htun ZT, Kim TS, Shin HJ, Na BK. Genetic polymorphism of merozoite surface protein-3 in Myanmar Plasmodium falciparum field isolates. Malar J 2020; 19:184. [PMID: 32429986 PMCID: PMC7235555 DOI: 10.1186/s12936-020-03256-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/08/2020] [Indexed: 11/10/2022] Open
Abstract
Background Plasmodium falciparum merozoite surface protein-3 (PfMSP-3) is a target of naturally acquired immunity against P. falciparum infection and is a promising vaccine candidate because of its critical role in the erythrocyte invasion of the parasite. Understanding the genetic diversity of pfmsp-3 is important for recognizing genetic nature and evolutionary aspect of the gene in the natural P. falciparum population and for designing an effective vaccine based on the antigen. Methods Blood samples collected from P. falciparum-infected patients in Naung Cho and Pyin Oo Lwin, Myanmar, in 2015 were used in this study. The pfmsp-3 was amplified by polymerase chain reaction, cloned, and sequenced. Genetic polymorphism and natural selection of Myanmar pfmsp-3 were analysed using the programs DNASTAR, MEGA6, and DnaSP 5.10.00. Genetic diversity and natural selection of the global pfmsp-3 were also comparatively analysed. Results Myanmar pfmsp-3 displayed 2 different alleles, 3D7 and K1. The 3D7 allelic type was predominant in the population, but genetic polymorphism was less diverse than for the K1 allelic type. Polymorphic characters in both allelic types were caused by amino acid substitutions, insertions, and deletions. Amino acid substitutions were mainly occurred at the alanine heptad repeat domains, whereas most insertions and deletions were found at the glutamate rich domain. Overall patterns of amino acid polymorphisms detected in Myanmar pfmsp-3 were similar in the global pfmsp-3 population, but novel amino acid changes were observed in Myanmar pfmsp-3 with low frequencies. Complicated patterns of natural selection and recombination events were predicted in the global pfmsp-3, which may act as major driving forces to maintain and generate genetic diversity of the global pfmsp-3 population. Conclusion Global pfmsp-3 revealed genetic polymorphisms, suggesting that the functional and structural consequences of the polymorphisms should be considered in designing a vaccine based on PfMSP-3. Further examination of genetic diversity of pfmsp-3 in the global P. falciparum population is necessary to gain in-depth insight for the population structure and evolutionary aspect of global pfmsp-3.
Collapse
Affiliation(s)
- Hương Giang Lê
- Department of Parasitology and Tropical Medicine, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea.,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Thị Lam Thái
- Department of Parasitology and Tropical Medicine, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea.,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Jung-Mi Kang
- Department of Parasitology and Tropical Medicine, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea.,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Jinyoung Lee
- Department of Tropical Medicine, Inha University College of Medicine, Incheon, 22212, Republic of Korea
| | - Mya Moe
- Department of Medical Research Pyin Oo Lwin Branch, Pyin Oo Lwin, Myanmar
| | - Tuấn Cường Võ
- Department of Parasitology and Tropical Medicine, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea.,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Haung Naw
- Department of Parasitology and Tropical Medicine, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea.,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Moe Kyaw Myint
- Department of Medical Research Pyin Oo Lwin Branch, Pyin Oo Lwin, Myanmar
| | - Zaw Than Htun
- Department of Medical Research Pyin Oo Lwin Branch, Pyin Oo Lwin, Myanmar
| | - Tong-Soo Kim
- Department of Tropical Medicine, Inha University College of Medicine, Incheon, 22212, Republic of Korea
| | - Ho-Joon Shin
- Department of Microbiology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Byoung-Kuk Na
- Department of Parasitology and Tropical Medicine, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea. .,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea.
| |
Collapse
|
22
|
Old and Recent Advances in Life Cycle, Pathogenesis, Diagnosis, Prevention, and Treatment of Malaria Including Perspectives in Ethiopia. ScientificWorldJournal 2020. [DOI: 10.1155/2020/1295381] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Malaria, caused by apicomplexan parasite, is an old disease and continues to be a major public health threat in many countries. This article aims to present different aspects of malaria including causes, pathogenesis, prevention, and treatment in an articulate and comprehensive manner. Six Plasmodium species are recognized as the etiology of human malaria, of which Plasmodium falciparum is popular in East and Southern Africa. Malaria is transmitted mainly through Anopheles gambiae and Anopheles funestus, the two most effective malaria vectors in the world. Half of the world’s population is at risk for malaria infection. Globally, the morbidity and mortality rates of malaria have become decreased even though few reports in Ethiopia showed high prevalence of malaria. The malaria parasite has a complex life cycle that takes place both inside the mosquito and human beings. Generally, diagnosis of malaria is classified into clinical and parasitological diagnoses. Lack of clear understanding on the overall biology of Plasmodium has created a challenge in an effort to develop new drugs, vaccines, and preventive methods against malaria. However, three types of vaccines and a lot of novel compounds are under perclinical and clinical studies that are triggered by the occurrence of resistance among commonly used drugs and insecticides. Antiadhesion adjunctive therapies are also under investigation in the laboratory. In addition to previously known targets for diagnostic tool, vaccine and drug discovery scientists from all corner of the world are in search of new targets and chemical entities.
Collapse
|
23
|
Narula AK, Azad CS, Nainwal LM. New dimensions in the field of antimalarial research against malaria resurgence. Eur J Med Chem 2019; 181:111353. [DOI: 10.1016/j.ejmech.2019.05.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 04/16/2019] [Accepted: 05/15/2019] [Indexed: 12/20/2022]
|
24
|
Dejon-Agobe JC, Ateba-Ngoa U, Lalremruata A, Homoet A, Engelhorn J, Nouatin OP, Edoa JR, Fernandes JF, Esen M, Mouwenda YD, Betouke Ongwe EM, Massinga-Loembe M, Hoffman SL, Sim BKL, Theisen M, Kremsner PG, Adegnika AA, Lell B, Mordmüller B. Controlled Human Malaria Infection of Healthy Adults With Lifelong Malaria Exposure to Assess Safety, Immunogenicity, and Efficacy of the Asexual Blood Stage Malaria Vaccine Candidate GMZ2. Clin Infect Dis 2019; 69:1377-1384. [PMID: 30561539 PMCID: PMC6763635 DOI: 10.1093/cid/ciy1087] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 12/18/2018] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND GMZ2 is a recombinant malaria vaccine inducing immune responses against Plasmodium falciparum (Pf) merozoite surface protein-3 and glutamate-rich protein. We used standardized controlled human malaria infection (CHMI) to assess the efficacy of this asexual blood-stage vaccine. METHODS We vaccinated 50 healthy, adult volunteers with lifelong exposure to Pf 3 times, at 4-week intervals, with 30 or 100 µg GMZ2 formulated in CAF01, a liposome-based adjuvant; 100 µg GMZ2, formulated in Alhydrogel; or a control vaccine (Verorab). Approximately 13 weeks after the last vaccination, 35/50 volunteers underwent CHMI by direct venous inoculation of 3200 Pf sporozoites (Sanaria® PfSPZ Challenge). RESULTS Adverse events were similarly distributed between GMZ2 and control vaccinees. Baseline-corrected anti-GMZ2 antibody concentrations 4 weeks after the last vaccination were higher in all 3 GMZ2-vaccinated arms, compared to the control group. All GMZ2 formulations induced similar antibody levels. CHMI resulted in 29/34 (85%) volunteers with Pf parasitemia and 15/34 (44%) with malaria (parasitemia and symptoms). The proportion of participants with malaria (2/5 control, 6/10 GMZ2-Alhydrogel, 2/8 30 µg GMZ2-CAF01, and 5/11 100 µg GMZ2-CAF01) and the time it took them to develop malaria were similar in all groups. Baseline, vaccine-specific antibody concentrations were associated with protection against malaria. CONCLUSIONS GMZ2 is well tolerated and immunogenic in lifelong-Pf-exposed adults from Gabon, with similar antibody responses regardless of formulation. CHMI showed no protective effect of prior vaccination with GMZ2, although baseline, vaccine-specific antibody concentrations were associated with protection. CHMI with the PfSPZ Challenge is a potent new tool to validate asexual, blood-stage malaria vaccines in Africa. CLINICAL TRIALS REGISTRATION Pan-African Clinical Trials: PACTR201503001038304.
Collapse
Affiliation(s)
- Jean Claude Dejon-Agobe
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Ulysse Ateba-Ngoa
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
- Department of Parasitology, Leiden University Medical Center, The Netherlands
| | - Albert Lalremruata
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Andreas Homoet
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Julie Engelhorn
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Odilon Paterne Nouatin
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Jean Ronald Edoa
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - José F Fernandes
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Meral Esen
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Yoanne Darelle Mouwenda
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Department of Parasitology, Leiden University Medical Center, The Netherlands
| | - Eunice M Betouke Ongwe
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Marguerite Massinga-Loembe
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | | | | | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Denmark
- Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Peter G Kremsner
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Ayôla A Adegnika
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
- Department of Parasitology, Leiden University Medical Center, The Netherlands
| | - Bertrand Lell
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Benjamin Mordmüller
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| |
Collapse
|
25
|
Odhiambo EO, Datta D, Guyah B, Ayodo G, Ondigo BN, Abong'o BO, John CC, Frosch AEP. HIV infection drives IgM and IgG3 subclass bias in Plasmodium falciparum-specific and total immunoglobulin concentration in Western Kenya. Malar J 2019; 18:297. [PMID: 31470903 PMCID: PMC6716850 DOI: 10.1186/s12936-019-2915-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 08/17/2019] [Indexed: 11/22/2022] Open
Abstract
Background HIV infection is associated with more frequent and severe episodes of malaria and may be the result of altered malaria-specific B cell responses. However, it is poorly understood how HIV and the associated lymphopenia and immune activation affect malaria-specific antibody responses. Methods HIV infected and uninfected adults were recruited from Bondo subcounty hospital in Western Kenya at the time of HIV testing (antiretroviral and co-trimoxazole prophylaxis naïve). Total and Plasmodium falciparum apical membrane antigen-1 (AMA1) and glutamate rich protein-R0 (GLURP-R0) specific IgM, IgG and IgG subclass concentrations was measured in 129 and 52 of recruited HIV-infected and uninfected individuals, respectively. In addition, HIV-1 viral load (VL), CD4+ T cell count, and C-reactive protein (CRP) concentration was quantified in study participants. Antibody levels were compared based on HIV status and the associations of antibody concentration with HIV-1 VL, CD4+ count, and CRP levels was measured using Spearman correlation testing. Results Among study participants, concentrations of IgM, IgG1 and IgG3 antibodies to AMA1 and GLURP-R0 were higher in HIV infected individuals compared to uninfected individuals (all p < 0.001). The IgG3 to IgG1 ratio to both AMA1 and GLURP-R0 was also significantly higher in HIV-infected individuals (p = 0.02). In HIV-infected participants, HIV-1 VL and CRP were weakly correlated with AMA1 and GLURP-R0 specific IgM and IgG1 concentrations and total (not antigen specific) IgM, IgG, IgG1, and IgG3 concentrations (all p < 0.05), suggesting that these changes are related in part to viral load and inflammation. Conclusions Overall, HIV infection leads to a total and malaria antigen-specific immunoglobulin production bias towards higher levels of IgM, IgG1, and IgG3, and HIV-1 viraemia and systemic inflammation are weakly correlated with these changes. Further assessments of antibody affinity and function and correlation with risk of clinical malaria, will help to better define the effects of HIV infection on clinical and biological immunity to malaria.
Collapse
Affiliation(s)
- Eliud O Odhiambo
- Department of Biomedical Science and Technology, Maseno University, Maseno, Kenya.,Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya.,Department of Pediatrics, Indiana University School of Medicine, Indianapolis, USA
| | - Dibyadyuti Datta
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, USA
| | - Bernard Guyah
- Department of Biomedical Science and Technology, Maseno University, Maseno, Kenya
| | - George Ayodo
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya.,Jaramogi Oginga Odinga University of Science and Technology, Bondo, Kenya
| | - Bartholomew N Ondigo
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya.,Department of Biochemistry and Molecular Biology, Egerton University, Nakuru, Kenya.,Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD, USA
| | - Benard O Abong'o
- Department of Biomedical Science and Technology, Maseno University, Maseno, Kenya.,Department of Biology, Faculty of Science and Technology, National University of Lesotho, Roma, Lesotho
| | - Chandy C John
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya.,Department of Pediatrics, Indiana University School of Medicine, Indianapolis, USA
| | - Anne E P Frosch
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya. .,Department of Medicine, University of Minnesota, Minneapolis, USA. .,Hennepin Healthcare Research Institute, Minneapolis, MN, USA.
| |
Collapse
|
26
|
Pham JV, Yilma MA, Feliz A, Majid MT, Maffetone N, Walker JR, Kim E, Cho HJ, Reynolds JM, Song MC, Park SR, Yoon YJ. A Review of the Microbial Production of Bioactive Natural Products and Biologics. Front Microbiol 2019; 10:1404. [PMID: 31281299 PMCID: PMC6596283 DOI: 10.3389/fmicb.2019.01404] [Citation(s) in RCA: 266] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 06/04/2019] [Indexed: 12/24/2022] Open
Abstract
A variety of organisms, such as bacteria, fungi, and plants, produce secondary metabolites, also known as natural products. Natural products have been a prolific source and an inspiration for numerous medical agents with widely divergent chemical structures and biological activities, including antimicrobial, immunosuppressive, anticancer, and anti-inflammatory activities, many of which have been developed as treatments and have potential therapeutic applications for human diseases. Aside from natural products, the recent development of recombinant DNA technology has sparked the development of a wide array of biopharmaceutical products, such as recombinant proteins, offering significant advances in treating a broad spectrum of medical illnesses and conditions. Herein, we will introduce the structures and diverse biological activities of natural products and recombinant proteins that have been exploited as valuable molecules in medicine, agriculture and insect control. In addition, we will explore past and ongoing efforts along with achievements in the development of robust and promising microorganisms as cell factories to produce biologically active molecules. Furthermore, we will review multi-disciplinary and comprehensive engineering approaches directed at improving yields of microbial production of natural products and proteins and generating novel molecules. Throughout this article, we will suggest ways in which microbial-derived biologically active molecular entities and their analogs could continue to inspire the development of new therapeutic agents in academia and industry.
Collapse
Affiliation(s)
- Janette V. Pham
- Geisinger Commonwealth School of Medicine, Scranton, PA, United States
- Baruch S. Blumberg Institute, Doylestown, PA, United States
| | - Mariamawit A. Yilma
- Geisinger Commonwealth School of Medicine, Scranton, PA, United States
- Baruch S. Blumberg Institute, Doylestown, PA, United States
| | - Adriana Feliz
- Geisinger Commonwealth School of Medicine, Scranton, PA, United States
- Baruch S. Blumberg Institute, Doylestown, PA, United States
| | - Murtadha T. Majid
- Geisinger Commonwealth School of Medicine, Scranton, PA, United States
- Baruch S. Blumberg Institute, Doylestown, PA, United States
| | - Nicholas Maffetone
- Geisinger Commonwealth School of Medicine, Scranton, PA, United States
- Baruch S. Blumberg Institute, Doylestown, PA, United States
| | - Jorge R. Walker
- Geisinger Commonwealth School of Medicine, Scranton, PA, United States
- Baruch S. Blumberg Institute, Doylestown, PA, United States
| | - Eunji Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, South Korea
| | - Hyo Je Cho
- School of Life Sciences and Biotechnology, Kyungpook National University, Daegu, South Korea
| | - Jared M. Reynolds
- Geisinger Commonwealth School of Medicine, Scranton, PA, United States
- Baruch S. Blumberg Institute, Doylestown, PA, United States
| | - Myoung Chong Song
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, South Korea
| | - Sung Ryeol Park
- Geisinger Commonwealth School of Medicine, Scranton, PA, United States
- Baruch S. Blumberg Institute, Doylestown, PA, United States
- Natural Products Discovery Institute, Doylestown, PA, United States
| | - Yeo Joon Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, South Korea
| |
Collapse
|
27
|
Wilson KL, Flanagan KL, Prakash MD, Plebanski M. Malaria vaccines in the eradication era: current status and future perspectives. Expert Rev Vaccines 2019; 18:133-151. [PMID: 30601095 DOI: 10.1080/14760584.2019.1561289] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The challenge to eradicate malaria is an enormous task that will not be achieved by current control measures, thus an efficacious and long-lasting malaria vaccine is required. The licensing of RTS, S/AS01 is a step forward in providing some protection, but a malaria vaccine that protects across multiple transmission seasons is still needed. To achieve this, inducing beneficial immune responses while minimising deleterious non-targeted effects will be essential. AREAS COVERED This article discusses the current challenges and advances in malaria vaccine development and reviews recent human clinical trials for each stage of infection. Pubmed and ScienceDirect were searched, focusing on cell mediated immunity and how T cell subsets might be targeted in future vaccines using novel adjuvants and emerging vaccine technologies. EXPERT COMMENTARY Despite decades of research there is no highly effective licensed malaria vaccine. However, there is cause for optimism as new adjuvants and vaccine systems emerge, and our understanding of correlates of protection increases, especially regarding cellular immunity. The new field of heterologous (non-specific) effects of vaccines also highlights the broader consequences of immunization. Importantly, the WHO led Malaria Vaccine Technology Roadmap illustrates that there is a political will among the global health community to make it happen.
Collapse
Affiliation(s)
- K L Wilson
- a Department of Immunology and Pathology, Faculty of Medicine, Nursing and Health Sciences , Monash University , Melbourne , Australia.,b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia
| | - K L Flanagan
- a Department of Immunology and Pathology, Faculty of Medicine, Nursing and Health Sciences , Monash University , Melbourne , Australia.,b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia.,c School of Medicine, Faculty of Health Sciences , University of Tasmania , Launceston , Australia
| | - M D Prakash
- b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia
| | - M Plebanski
- b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia
| |
Collapse
|
28
|
Beeson JG, Kurtovic L, Dobaño C, Opi DH, Chan JA, Feng G, Good MF, Reiling L, Boyle MJ. Challenges and strategies for developing efficacious and long-lasting malaria vaccines. Sci Transl Med 2019; 11:11/474/eaau1458. [DOI: 10.1126/scitranslmed.aau1458] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/05/2018] [Accepted: 11/02/2018] [Indexed: 12/24/2022]
Abstract
Although there has been major recent progress in malaria vaccine development, substantial challenges remain for achieving highly efficacious and durable vaccines against Plasmodium falciparum and Plasmodium vivax malaria. Greater knowledge of mechanisms and key targets of immunity are needed to accomplish this goal, together with new strategies for generating potent, long-lasting, functional immunity against multiple antigens. Implementation considerations in endemic areas will ultimately affect vaccine effectiveness, so innovations to simplify and enhance delivery are also needed. Whereas challenges remain, recent exciting progress and emerging knowledge promise hope for the future of malaria vaccines.
Collapse
|
29
|
Quintana MDP, Ch’ng JH, Zandian A, Imam M, Hultenby K, Theisen M, Nilsson P, Qundos U, Moll K, Chan S, Wahlgren M. SURGE complex of Plasmodium falciparum in the rhoptry-neck (SURFIN4.2-RON4-GLURP) contributes to merozoite invasion. PLoS One 2018; 13:e0201669. [PMID: 30092030 PMCID: PMC6084945 DOI: 10.1371/journal.pone.0201669] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/19/2018] [Indexed: 12/25/2022] Open
Abstract
Plasmodium falciparum invasion into red blood cells (RBCs) is a complex process engaging proteins on the merozoite surface and those contained and sequentially released from the apical organelles (micronemes and rhoptries). Fundamental to invasion is the formation of a moving junction (MJ), a region of close apposition of the merozoite and the RBC plasma membranes, through which the merozoite draws itself before settling into a newly formed parasitophorous vacuole (PV). SURFIN4.2 was identified at the surface of the parasitized RBCs (pRBCs) but was also found apically associated with the merozoite. Using antibodies against the N-terminus of the protein we show the presence of SURFIN4.2 in the neck of the rhoptries, its secretion into the PV and shedding into the culture supernatant upon schizont rupture. Using immunoprecipitation followed by mass spectrometry we describe here a novel protein complex we have named SURGE where SURFIN4.2 forms interacts with the rhoptry neck protein 4 (RON4) and the Glutamate Rich Protein (GLURP). The N-terminal cysteine-rich-domain (CRD) of SURFIN4.2 mediates binding to the RBC membrane and its interaction with RON4 suggests its involvement in the contact between the merozoite apex and the RBC at the MJ. Supporting this suggestion, we also found that polyclonal antibodies to the extracellular domain (including the CRD) of SURFIN4.2 partially inhibit merozoite invasion. We propose that the formation of the SURGE complex participates in the establishment of parasite infection within the PV and the RBCs.
Collapse
Affiliation(s)
- Maria del Pilar Quintana
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, Stockholm, Sweden
| | - Jun-Hong Ch’ng
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, Stockholm, Sweden
- Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Arash Zandian
- Affinity Proteomics, Science for Life Laboratory, School of Biotechnology, KTH-Royal Institutet of Technology, Stockholm, Sweden
| | - Maryam Imam
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, Stockholm, Sweden
| | - Kjell Hultenby
- Division of Clinical Research Centre, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Centre for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Peter Nilsson
- Affinity Proteomics, Science for Life Laboratory, School of Biotechnology, KTH-Royal Institutet of Technology, Stockholm, Sweden
| | - Ulrika Qundos
- Affinity Proteomics, Science for Life Laboratory, School of Biotechnology, KTH-Royal Institutet of Technology, Stockholm, Sweden
| | - Kirsten Moll
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, Stockholm, Sweden
| | - Sherwin Chan
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, Stockholm, Sweden
| | - Mats Wahlgren
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
30
|
Pattaradilokrat S, Trakoolsoontorn C, Simpalipan P, Warrit N, Kaewthamasorn M, Harnyuttanakorn P. Size and sequence polymorphisms in the glutamate-rich protein gene of the human malaria parasite Plasmodium falciparum in Thailand. Parasit Vectors 2018; 11:49. [PMID: 29357909 PMCID: PMC5778735 DOI: 10.1186/s13071-018-2630-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/08/2018] [Indexed: 11/10/2022] Open
Abstract
Background The glutamate-rich protein (GLURP) of the malaria parasite Plasmodium falciparum is a key surface antigen that serves as a component of a clinical vaccine. Moreover, the GLURP gene is also employed routinely as a genetic marker for malarial genotyping in epidemiological studies. While extensive size polymorphisms in GLURP are well recorded, the extent of the sequence diversity of this gene is rarely investigated. The present study aimed to explore the genetic diversity of GLURP in natural populations of P. falciparum. Results The polymorphic C-terminal repetitive R2 region of GLURP sequences from 65 P. falciparum isolates in Thailand were generated and combined with the data from 103 worldwide isolates to generate a GLURP database. The collection was comprised of 168 alleles, encoding 105 unique GLURP subtypes, characterized by 18 types of amino acid repeat units (AAU). Of these, 28 GLURP subtypes, formed by 10 AAU types, were detected in P. falciparum in Thailand. Among them, 19 GLURP subtypes and 2 AAU types are described for the first time in the Thai parasite population. The AAU sequences were highly conserved, which is likely due to negative selection. Standard Fst analysis revealed the shared distributions of GLURP types among the P. falciparum populations, providing evidence of gene flow among the different demographic populations. Conclusions Sequence diversity causing size variations in GLURP in Thai P. falciparum populations were detected, and caused by non-synonymous substitutions in repeat units and some insertion/deletion of aspartic acid or glutamic acid codons between repeat units. The P. falciparum population structure based on GLURP showed promising implications for the development of GLURP-based vaccines and for monitoring vaccine efficacy. Electronic supplementary material The online version of this article (doi: 10.1186/s13071-018-2630-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sittiporn Pattaradilokrat
- Department of Biology, Faculty of Science, Chualongkorn University, 254 Phayathai Road, Bangkok, 10330, Thailand. .,Veterinary Parasitology Research Group, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Chawinya Trakoolsoontorn
- Department of Biology, Faculty of Science, Chualongkorn University, 254 Phayathai Road, Bangkok, 10330, Thailand
| | - Phumin Simpalipan
- Department of Biology, Faculty of Science, Chualongkorn University, 254 Phayathai Road, Bangkok, 10330, Thailand
| | - Natapot Warrit
- Department of Biology, Faculty of Science, Chualongkorn University, 254 Phayathai Road, Bangkok, 10330, Thailand
| | - Morakot Kaewthamasorn
- Veterinary Parasitology Research Group, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Pongchai Harnyuttanakorn
- Department of Biology, Faculty of Science, Chualongkorn University, 254 Phayathai Road, Bangkok, 10330, Thailand
| |
Collapse
|
31
|
Theisen M, Adu B, Mordmüller B, Singh S. The GMZ2 malaria vaccine: from concept to efficacy in humans. Expert Rev Vaccines 2017; 16:907-917. [PMID: 28699823 DOI: 10.1080/14760584.2017.1355246] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION GMZ2 is a recombinant protein consisting of conserved domains of GLURP and MSP3, two asexual blood-stage antigens of Plasmodium falciparum, and is designed with the aim of mimicking naturally acquired anti-malarial immunity. The rationale for combining these two antigens is based on a series of immune epidemiological studies from geographically diverse malaria endemic regions; functional in vitro studies; and pre-clinical studies in rodents and New World monkeys. GMZ2 adjuvanted with alhydrogel® (alum) was well tolerated and immunogenic in three phase 1 studies. The recently concluded phase 2 trial of GMZ2/alum, involving 1849 participants 12 to 60 month of age in four countries in West, Central and Eastern Africa, showed that GMZ2 is well tolerated and has some, albeit modest, efficacy in the target population. Areas covered: PubMed ( www.ncbi.nlm.nih.gov/pubmed ) was searched to review the progress and future prospects for clinical development of GMZ2 sub-unit vaccine. We will focus on discovery, naturally acquired immunity, functional activity of specific antibodies, sequence diversity, production, pre-clinical and clinical studies. Expert commentary: GMZ2 is well tolerated and has some, albeit modest, efficacy in the target population. More immunogenic formulations should be developed.
Collapse
Affiliation(s)
- Michael Theisen
- a Department for Congenital Disorders , Statens Serum Institut , Copenhagen , Denmark.,b Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology , University of Copenhagen , Copenhagen , Denmark.,c Department of Infectious Diseases , Copenhagen University Hospital , Rigshospitalet , Denmark
| | - Bright Adu
- d Noguchi Memorial Institute for Medical Research , University of Ghana , Legon , Ghana
| | - Benjamin Mordmüller
- e Institute of Tropical Medicine and Center for Infection Research, partner site Tübingen , University of Tübingen , Tübingen , Germany
| | - Subhash Singh
- f Indian Institute of Integrative Medicine , Jammu , India
| |
Collapse
|
32
|
Hill DL, Schofield L, Wilson DW. IgG opsonization of merozoites: multiple immune mechanisms for malaria vaccine development. Int J Parasitol 2017; 47:585-595. [PMID: 28668325 DOI: 10.1016/j.ijpara.2017.05.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 04/12/2017] [Accepted: 05/12/2017] [Indexed: 02/07/2023]
Abstract
Global eradication of the human-infecting malaria parasite Plasmodium falciparum, the major cause of malaria mortality, is unlikely to be achieved without an effective vaccine. However, our limited understanding of how protective immune responses target malaria parasites in humans, and how to best elicit these immune responses through vaccination, has hampered vaccine development. The red blood cell invading stage of the parasite lifecycle (merozoite) displays antigens that are attractive vaccine candidates as they are accessible to antibodies and raise high antibody titres in naturally immune individuals. The number of merozoite antigens that elicit an immune response, and their structural and functional diversity, has led to a large number of lead antigens being pursued as vaccine candidates. Despite being seemingly spoilt for choice in terms of vaccine candidates, there is still a lack of consensus on exactly how merozoite antibodies reduce parasitemia and malaria disease. In this review we describe the various immune mechanisms that can result from IgG opsonization of merozoites, and highlight recent developments that support a role for these functional antibodies in naturally acquired and vaccine-induced immunity.
Collapse
Affiliation(s)
- Danika L Hill
- Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom; The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia.
| | - Louis Schofield
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Australia
| | - Danny W Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia; Burnet Institute, 85 Commercial Road, Melbourne 3004, Victoria, Australia.
| |
Collapse
|
33
|
Kana IH, Adu B, Tiendrebeogo RW, Singh SK, Dodoo D, Theisen M. Naturally Acquired Antibodies Target the Glutamate-Rich Protein on Intact Merozoites and Predict Protection Against Febrile Malaria. J Infect Dis 2017; 215:623-630. [PMID: 28329101 DOI: 10.1093/infdis/jiw617] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/05/2017] [Indexed: 11/14/2022] Open
Abstract
Background Plasmodium species antigens accessible at the time of merozoite release are likely targets of biologically functional antibodies. Methods Immunoglobulin G (IgG) antibodies against intact merozoites were quantified in the plasma of Ghanaian children from a longitudinal cohort using a novel flow cytometry-based immunofluorescence assay. Functionality of these antibodies, as well as glutamate-rich protein (GLURP)-specific affinity-purified IgG from malaria hyperimmune Liberian adults, was assessed by the opsonic phagocytosis (OP) assay. Results Opsonic phagocytosis activity was strongly associated (hazard ratio [HR] = 0.46; 95% confidence interval [CI] = .30-.73; P = .0008) with protection against febrile malaria. Of the antimerozoite-specific antibodies, only IgG3 was significantly associated with both OP and protection (HR = 0.53; 95% CI = .34-.84; Pcorrected = .03) against febrile malaria. Similarly, GLURP-specific antibodies previously shown to be protective against febrile malaria in this same cohort were significantly associated with OP activity in this study. GLURP-specific antibodies recognized merozoites and also mediated OP activity. Conclusions These findings support previous studies that found OP of merozoites to be associated with protection against malaria and further shows IgG3 and GLURP antibodies are key in the OP mechanism, thus giving further impetus for the development of malaria vaccines targeting GLURP.
Collapse
Affiliation(s)
- Ikhlaq Hussain Kana
- Department for Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen.,Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Bright Adu
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon
| | - Régis Wendpayangde Tiendrebeogo
- Department for Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen.,Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Susheel Kumar Singh
- Department for Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen.,Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Daniel Dodoo
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen.,Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
34
|
Milligan P, Flach C, Theisen M. Efficacy of the GMZ2 malaria vaccine in African children. Vaccine 2017; 35:202. [DOI: 10.1016/j.vaccine.2016.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 11/09/2016] [Indexed: 11/30/2022]
|
35
|
Mishra M, Mishra VK, Kashaw V, Iyer AK, Kashaw SK. Comprehensive review on various strategies for antimalarial drug discovery. Eur J Med Chem 2016; 125:1300-1320. [PMID: 27886547 DOI: 10.1016/j.ejmech.2016.11.025] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/07/2016] [Accepted: 11/11/2016] [Indexed: 01/14/2023]
Abstract
The resistance of malaria parasites to existing drugs carries on growing and progressively limiting our ability to manage this severe disease and finally lead to a massive global health burden. Till now, malaria control has relied upon the traditional quinoline, antifolate and artemisinin compounds. Very few new antimalarials were developed in the past 50 years. Among recent approaches, identification of novel chemotherapeutic targets, exploration of natural products with medicinal significance, covalent bitherapy having a dual mode of action into a single hybrid molecule and malaria vaccine development are explored heavily. The proper execution of these approaches and proper investment from international agencies will accelerate the discovery of drugs that provide new hope for the control or eventual eradication of this global infectious disease. This review explores various strategies for assessment and development of new antimalarial drugs. Current status and scientific value of previous approaches are systematically reviewed and new approaches provide a pragmatic forecast for future developments are introduced as well.
Collapse
Affiliation(s)
- Mitali Mishra
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar, MP, India
| | - Vikash K Mishra
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar, MP, India
| | - Varsha Kashaw
- SVN Institute of Pharmaceutical Sciences, SVN University, Sagar, MP, India
| | - Arun K Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA
| | - Sushil Kumar Kashaw
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar, MP, India; Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
36
|
Pattaradilokrat S, Sawaswong V, Simpalipan P, Kaewthamasorn M, Siripoon N, Harnyuttanakorn P. Genetic diversity of the merozoite surface protein-3 gene in Plasmodium falciparum populations in Thailand. Malar J 2016; 15:517. [PMID: 27769257 PMCID: PMC5073822 DOI: 10.1186/s12936-016-1566-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 10/07/2016] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND An effective malaria vaccine is an urgently needed tool to fight against human malaria, the most deadly parasitic disease of humans. One promising candidate is the merozoite surface protein-3 (MSP-3) of Plasmodium falciparum. This antigenic protein, encoded by the merozoite surface protein (msp-3) gene, is polymorphic and classified according to size into the two allelic types of K1 and 3D7. A recent study revealed that both the K1 and 3D7 alleles co-circulated within P. falciparum populations in Thailand, but the extent of the sequence diversity and variation within each allelic type remains largely unknown. METHODS The msp-3 gene was sequenced from 59 P. falciparum samples collected from five endemic areas (Mae Hong Son, Kanchanaburi, Ranong, Trat and Ubon Ratchathani) in Thailand and analysed for nucleotide sequence diversity, haplotype diversity and deduced amino acid sequence diversity. The gene was also subject to population genetic analysis (F st ) and neutrality tests (Tajima's D, Fu and Li D* and Fu and Li' F* tests) to determine any signature of selection. RESULTS The sequence analyses revealed eight unique DNA haplotypes and seven amino acid sequence variants, with a haplotype and nucleotide diversity of 0.828 and 0.049, respectively. Neutrality tests indicated that the polymorphism detected in the alanine heptad repeat region of MSP-3 was maintained by positive diversifying selection, suggesting its role as a potential target of protective immune responses and supporting its role as a vaccine candidate. Comparison of MSP-3 variants among parasite populations in Thailand, India and Nigeria also inferred a close genetic relationship between P. falciparum populations in Asia. CONCLUSION This study revealed the extent of the msp-3 gene diversity in P. falciparum in Thailand, providing the fundamental basis for the better design of future blood stage malaria vaccines against P. falciparum.
Collapse
Affiliation(s)
| | - Vorthon Sawaswong
- Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Phumin Simpalipan
- Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Morakot Kaewthamasorn
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Napaporn Siripoon
- College of Public Health Sciences, Chulalongkorn University, Bangkok, 10330 Thailand
| | | |
Collapse
|
37
|
A phase 2b randomized, controlled trial of the efficacy of the GMZ2 malaria vaccine in African children. Vaccine 2016; 34:4536-4542. [PMID: 27477844 DOI: 10.1016/j.vaccine.2016.07.041] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/07/2016] [Accepted: 07/20/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND GMZ2 is a recombinant protein malaria vaccine, comprising two blood-stage antigens of Plasmodium falciparum, glutamate-rich protein and merozoite surface protein 3. We assessed efficacy of GMZ2 in children in Burkina Faso, Gabon, Ghana and Uganda. METHODS Children 12-60months old were randomized to receive three injections of either 100μg GMZ2 adjuvanted with aluminum hydroxide or a control vaccine (rabies) four weeks apart and were followed up for six months to measure the incidence of malaria defined as fever or history of fever and a parasite density ⩾5000/μL. RESULTS A cohort of 1849 children were randomized, 1735 received three doses of vaccine (868 GMZ2, 867 control-vaccine). There were 641 malaria episodes in the GMZ2/Alum group and 720 in the control group. In the ATP analysis, vaccine efficacy (VE), adjusted for age and site was 14% (95% confidence interval [CI]: 3.6%, 23%, p-value=0.009). In the ITT analysis, age-adjusted VE was 11.3% (95% CI 2.5%, 19%, p-value=0.013). VE was higher in older children. In GMZ2-vaccinated children, the incidence of malaria decreased with increasing vaccine-induced anti-GMZ2 IgG concentration. There were 32 cases of severe malaria (18 in the rabies vaccine group and 14 in the GMZ2 group), VE 27% (95% CI -44%, 63%). CONCLUSIONS GMZ2 is the first blood-stage malaria vaccine to be evaluated in a large multicenter trial. GMZ2 was well tolerated and immunogenic, and reduced the incidence of malaria, but efficacy would need to be substantially improved, using a more immunogenic formulation, for the vaccine to have a public health role.
Collapse
|
38
|
Boyle MJ, Reiling L, Osier FH, Fowkes FJI. Recent insights into humoral immunity targeting Plasmodium falciparum and Plasmodium vivax malaria. Int J Parasitol 2016; 47:99-104. [PMID: 27451359 DOI: 10.1016/j.ijpara.2016.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/08/2016] [Accepted: 06/09/2016] [Indexed: 01/02/2023]
Abstract
Recent efforts in malaria control have led to marked reductions in malaria incidence. However, new strategies are needed to sustain malaria elimination and eradication and achieve the World Health Organization goal of a malaria-free world. The development of highly effective vaccines would contribute to this goal and would be facilitated by a comprehensive understanding of humoral immune responses targeting Plasmodium falciparum and Plasmodium vivax malaria. New tools are required to facilitate the identification of vaccine candidates and the development of vaccines that induce functional and protective immunity. Here we discuss recent published findings, and unpublished work presented at the 2016 Molecular Approaches to Malaria conference, that highlight advancements in understanding humoral immune responses in the context of vaccine development. Highlights include the increased application of 'omics' and 'Big data' platforms to identify vaccine candidates, and the identification of novel functions of antibody responses that mediate protection. The application of these strategies and a global approach will increase the likelihood of rapid development of highly efficacious vaccines.
Collapse
Affiliation(s)
- Michelle J Boyle
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria 3004, Australia; Menzies School of Medical Research, Darwin, Northern Territory 0810, Australia.
| | - Linda Reiling
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria 3004, Australia
| | - Faith H Osier
- KEMRI Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | - Freya J I Fowkes
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria 3004, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria 3010, Australia; Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia; Department of Infectious Diseases, Monash University, Melbourne, Victoria 3004, Australia
| |
Collapse
|
39
|
Favuzza P, Blaser S, Dreyer AM, Riccio G, Tamborrini M, Thoma R, Matile H, Pluschke G. Generation of Plasmodium falciparum parasite-inhibitory antibodies by immunization with recombinantly-expressed CyRPA. Malar J 2016; 15:161. [PMID: 26979066 PMCID: PMC4791974 DOI: 10.1186/s12936-016-1213-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 03/05/2016] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The pathogenesis of malaria is primarily associated with blood-stage infection and there is strong evidence that antibodies specific for parasite blood-stage antigens can control parasitaemia. This provides a strong rationale for incorporation of asexual blood-stage antigen components into an effective multivalent malaria subunit vaccine. On the basis of available genome-wide transcriptomic and proteomic data, previously uncharacterized Plasmodium falciparum open reading frames were screened for new blood stage vaccine candidates. This has led to the identification of the cysteine-rich protective antigen (PfCyRPA), which forms together with PfRH5 and PfRipr a multiprotein complex that is crucial for erythrocyte invasion. METHODS Glycosylated and non-glycosylated variants of recombinant PfCyRPA were expressed and produced as secreted protein in mammalian cells. Adjuvanted formulations of purified PfCyRPA were tested to assess whether they can effectively elicit parasite inhibitory antibodies, and to investigate whether or not the glycosylation status affects antibody binding. For this purpose, two sets of PfCyRPA-specific mouse monoclonal antibodies (mAbs) have been raised and evaluated for functional activity. RESULTS Generated PfCyRPA-specific mAbs, irrespective of the immunogen's glycosylation status, showed substantial parasite in vitro growth-inhibitory activity due to inhibition of erythrocyte invasion by merozoites. Furthermore, passive immunization experiments in P. falciparum infected NOD-scid IL2Rγ (null) mice engrafted with human erythrocytes demonstrated potent in vivo growth-inhibitory activity of generated mAbs. CONCLUSIONS Recombinantly expressed PfCyRPA tested as adjuvanted vaccine formulations in mice elicited antibodies that significantly inhibit P. falciparum asexual blood stage parasite growth both in vitro and in vivo. These findings render PfCyRPA a promising blood-stage candidate antigen for inclusion into a multicomponent malaria subunit vaccine.
Collapse
Affiliation(s)
- Paola Favuzza
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Simon Blaser
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Anita M Dreyer
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Guy Riccio
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Marco Tamborrini
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Ralf Thoma
- Roche Pharmaceutical Research & Early Development, Small Molecule Research, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Hugues Matile
- Roche Pharmaceutical Research & Early Development, Small Molecule Research, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Gerd Pluschke
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland. .,University of Basel, Basel, Switzerland.
| |
Collapse
|
40
|
Drinkwater N, Vinh NB, Mistry SN, Bamert RS, Ruggeri C, Holleran JP, Loganathan S, Paiardini A, Charman SA, Powell AK, Avery VM, McGowan S, Scammells PJ. Potent dual inhibitors of Plasmodium falciparum M1 and M17 aminopeptidases through optimization of S1 pocket interactions. Eur J Med Chem 2016; 110:43-64. [DOI: 10.1016/j.ejmech.2016.01.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/11/2016] [Accepted: 01/11/2016] [Indexed: 10/22/2022]
|
41
|
Abstract
There have been significant decreases in malaria mortality and morbidity in the last 10-15 years, and the most advanced pre-erythrocytic malaria vaccine, RTS,S, received a positive opinion from European regulators in July 2015. However, no blood-stage vaccine has reached a phase III trial. The first part of this review summarizes the pros and cons of various assays and models that have been and will be used to predict the efficacy of blood-stage vaccines. In the second part, blood-stage vaccine candidates that showed some efficacy in human clinical trials or controlled human malaria infection models are discussed. Then, candidates under clinical investigation are described in the third part, and other novel candidates and strategies are reviewed in the last part.
Collapse
Affiliation(s)
- Kazutoyo Miura
- a Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases , National Institutes of Health , Rockville , MD , USA
| |
Collapse
|
42
|
Beeson JG, Drew DR, Boyle MJ, Feng G, Fowkes FJI, Richards JS. Merozoite surface proteins in red blood cell invasion, immunity and vaccines against malaria. FEMS Microbiol Rev 2016; 40:343-72. [PMID: 26833236 PMCID: PMC4852283 DOI: 10.1093/femsre/fuw001] [Citation(s) in RCA: 242] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2016] [Indexed: 01/11/2023] Open
Abstract
Malaria accounts for an enormous burden of disease globally, with Plasmodium falciparum accounting for the majority of malaria, and P. vivax being a second important cause, especially in Asia, the Americas and the Pacific. During infection with Plasmodium spp., the merozoite form of the parasite invades red blood cells and replicates inside them. It is during the blood-stage of infection that malaria disease occurs and, therefore, understanding merozoite invasion, host immune responses to merozoite surface antigens, and targeting merozoite surface proteins and invasion ligands by novel vaccines and therapeutics have been important areas of research. Merozoite invasion involves multiple interactions and events, and substantial processing of merozoite surface proteins occurs before, during and after invasion. The merozoite surface is highly complex, presenting a multitude of antigens to the immune system. This complexity has proved challenging to our efforts to understand merozoite invasion and malaria immunity, and to developing merozoite antigens as malaria vaccines. In recent years, there has been major progress in this field, and several merozoite surface proteins show strong potential as malaria vaccines. Our current knowledge on this topic is reviewed, highlighting recent advances and research priorities. The authors summarize current knowledge of merozoite surface proteins of malaria parasites; their function in invasion, processing of surface proteins before, during and after invasion, their importance as targets of immunity, and the current status of malaria vaccines that target merozoite surface proteins.
Collapse
Affiliation(s)
- James G Beeson
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia Department of Microbiology, Monash University, Clayton, Victoria, Australia Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Damien R Drew
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia
| | - Michelle J Boyle
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia
| | - Gaoqian Feng
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia
| | - Freya J I Fowkes
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia Department of Epidemiology and Preventive Medicine, Monash University, Clayton, Victoria, Australia School of Population Health, University of Melbourne, Parkville, Victoria, Australia
| | - Jack S Richards
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia Department of Microbiology, Monash University, Clayton, Victoria, Australia Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
43
|
Draper SJ, Angov E, Horii T, Miller LH, Srinivasan P, Theisen M, Biswas S. Recent advances in recombinant protein-based malaria vaccines. Vaccine 2015; 33:7433-43. [PMID: 26458807 PMCID: PMC4687528 DOI: 10.1016/j.vaccine.2015.09.093] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 09/05/2015] [Accepted: 09/28/2015] [Indexed: 01/03/2023]
Abstract
Protein-based vaccines remain the cornerstone approach for B cell and antibody induction against leading target malaria antigens. Advances in antigen selection, immunogen design and epitope-focusing are advancing the field. New heterologous expression platforms are enabling cGMP production of next-generation protein vaccines. Next-generation antigens, protein-based immunogens and virus-like particle (VLP) delivery platforms are in clinical development. Protein-based vaccines will form part of a highly effective multi-component/multi-stage/multi-antigen subunit formulation against malaria.
Plasmodium parasites are the causative agent of human malaria, and the development of a highly effective vaccine against infection, disease and transmission remains a key priority. It is widely established that multiple stages of the parasite's complex lifecycle within the human host and mosquito vector are susceptible to vaccine-induced antibodies. The mainstay approach to antibody induction by subunit vaccination has been the delivery of protein antigen formulated in adjuvant. Extensive efforts have been made in this endeavor with respect to malaria vaccine development, especially with regard to target antigen discovery, protein expression platforms, adjuvant testing, and development of soluble and virus-like particle (VLP) delivery platforms. The breadth of approaches to protein-based vaccines is continuing to expand as innovative new concepts in next-generation subunit design are explored, with the prospects for the development of a highly effective multi-component/multi-stage/multi-antigen formulation seeming ever more likely. This review will focus on recent progress in protein vaccine design, development and/or clinical testing for a number of leading malaria antigens from the sporozoite-, merozoite- and sexual-stages of the parasite's lifecycle–including PfCelTOS, PfMSP1, PfAMA1, PfRH5, PfSERA5, PfGLURP, PfMSP3, Pfs48/45 and Pfs25. Future prospects and challenges for the development, production, human delivery and assessment of protein-based malaria vaccines are discussed.
Collapse
Affiliation(s)
- Simon J Draper
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK.
| | - Evelina Angov
- Walter Reed Army Institute of Research, U. S. Military Malaria Research Program, Malaria Vaccine Branch, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - Toshihiro Horii
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 561-873, Japan
| | - Louis H Miller
- Malaria Cell Biology Section, Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Prakash Srinivasan
- Malaria Cell Biology Section, Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark; Centre for Medical Parasitology at Department of International Health, Immunology, and Microbiology and Department of Infectious Diseases, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sumi Biswas
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| |
Collapse
|
44
|
Llewellyn D, Miura K, Fay MP, Williams AR, Murungi LM, Shi J, Hodgson SH, Douglas AD, Osier FH, Fairhurst RM, Diakite M, Pleass RJ, Long CA, Draper SJ. Standardization of the antibody-dependent respiratory burst assay with human neutrophils and Plasmodium falciparum malaria. Sci Rep 2015; 5:14081. [PMID: 26373337 PMCID: PMC4571651 DOI: 10.1038/srep14081] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 08/17/2015] [Indexed: 01/09/2023] Open
Abstract
The assessment of naturally-acquired and vaccine-induced immunity to blood-stage Plasmodium falciparum malaria is of long-standing interest. However, the field has suffered from a paucity of in vitro assays that reproducibly measure the anti-parasitic activity induced by antibodies in conjunction with immune cells. Here we optimize the antibody-dependent respiratory burst (ADRB) assay, which assesses the ability of antibodies to activate the release of reactive oxygen species from human neutrophils in response to P. falciparum blood-stage parasites. We focus particularly on assay parameters affecting serum preparation and concentration, and importantly assess reproducibility. Our standardized protocol involves testing each serum sample in singlicate with three independent neutrophil donors, and indexing responses against a standard positive control of pooled hyper-immune Kenyan sera. The protocol can be used to quickly screen large cohorts of samples from individuals enrolled in immuno-epidemiological studies or clinical vaccine trials, and requires only 6 μL of serum per sample. Using a cohort of 86 samples, we show that malaria-exposed individuals induce higher ADRB activity than malaria-naïve individuals. The development of the ADRB assay complements the use of cell-independent assays in blood-stage malaria, such as the assay of growth inhibitory activity, and provides an important standardized cell-based assay in the field.
Collapse
Affiliation(s)
- David Llewellyn
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Michael P. Fay
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Andrew R. Williams
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Linda M. Murungi
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
- KEMRI Centre for Geographic Medicine Research, Coast, P.O. Box 230-80108, Kilifi, Kenya
| | - Jianguo Shi
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Susanne H. Hodgson
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Alexander D. Douglas
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Faith H. Osier
- KEMRI Centre for Geographic Medicine Research, Coast, P.O. Box 230-80108, Kilifi, Kenya
| | - Rick M. Fairhurst
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Mahamadou Diakite
- Malaria Research and Training Centre, Faculty of Medicine, Pharmacy and Odonto-stomatology, University of Bamako, Bamako, Mali
| | - Richard J. Pleass
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Carole A. Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Simon J. Draper
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| |
Collapse
|
45
|
Moreno A, Joyner C. Malaria vaccine clinical trials: what's on the horizon. Curr Opin Immunol 2015; 35:98-106. [PMID: 26172291 DOI: 10.1016/j.coi.2015.06.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 06/12/2015] [Accepted: 06/22/2015] [Indexed: 01/01/2023]
Abstract
Significant progress toward a malaria vaccine, specifically for Plasmodium falciparum, has been made in the past few years with the completion of numerous clinical trials. Each trial has utilized a unique combination of antigens, delivery platforms, and adjuvants, which has provided the research community with a wealth of critical information to apply towards the development of next generation malaria vaccines. Despite the progress toward a P. falciparum vaccine, P. vivax vaccine research requires more momentum and additional investigations to identify novel vaccine candidates. In this review, recently completed and ongoing malaria vaccine clinical trials as well as vaccine candidates that are in the development pipeline are reviewed. Perspectives for future research using post-genomic mining, nonhuman primate models, and systems biology are also discussed.
Collapse
Affiliation(s)
- Alberto Moreno
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA 30329, USA; Malaria Host-Pathogen Interaction Center, Emory University, 954 Gatewood Road, Atlanta, GA 30329, USA; Division of Infectious Diseases, Department of Medicine, Emory University, 69 Jesse Hill, Jr. Drive, SE, Atlanta, GA 30303, USA.
| | - Chester Joyner
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA 30329, USA; Malaria Host-Pathogen Interaction Center, Emory University, 954 Gatewood Road, Atlanta, GA 30329, USA
| |
Collapse
|
46
|
Structural basis for epitope masking and strain specificity of a conserved epitope in an intrinsically disordered malaria vaccine candidate. Sci Rep 2015; 5:10103. [PMID: 25965408 PMCID: PMC4428071 DOI: 10.1038/srep10103] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/30/2015] [Indexed: 12/15/2022] Open
Abstract
Merozoite surface protein 2 (MSP2) is an intrinsically disordered, membrane-anchored antigen of the malaria parasite Plasmodium falciparum. MSP2 can elicit a protective, albeit strain-specific, antibody response in humans. Antibodies are generated to the conserved N- and C-terminal regions but many of these react poorly with the native antigen on the parasite surface. Here we demonstrate that recognition of a conserved N-terminal epitope by mAb 6D8 is incompatible with the membrane-bound conformation of that region, suggesting a mechanism by which native MSP2 escapes antibody recognition. Furthermore, crystal structures and NMR spectroscopy identify transient, strain-specific interactions between the 6D8 antibody and regions of MSP2 beyond the conserved epitope. These interactions account for the differential affinity of 6D8 for the two allelic families of MSP2, even though 6D8 binds to a fully conserved epitope. These results highlight unappreciated mechanisms that may modulate the specificity and efficacy of immune responses towards disordered antigens.
Collapse
|
47
|
Kaddumukasa M, Lwanira C, Lugaajju A, Katabira E, Persson KEM, Wahlgren M, Kironde F. Parasite Specific Antibody Increase Induced by an Episode of Acute P. falciparum Uncomplicated Malaria. PLoS One 2015; 10:e0124297. [PMID: 25906165 PMCID: PMC4408068 DOI: 10.1371/journal.pone.0124297] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 03/11/2015] [Indexed: 11/30/2022] Open
Abstract
Introduction There is no approved vaccine for malaria, and precisely how human antibody responses to malaria parasite components and potential vaccine molecules are developed and maintained remains poorly defined. In this study, antibody anamnestic or memory response elicited by a single episode of P. falciparum infection was investigated. Methods This study involved 362 malaria patients aged between 6 months to 60 years, of whom 19% were early-diagnosed people living with HIV/AIDS (PLWHA). On the day malaria was diagnosed and 42 days later, blood specimens were collected. Parasite density, CD4+ cells, and antibodies specific to synthetic peptides representing antigenic regions of the P. falciparum proteins GLURP, MSP3 and HRPII were measured. Results On the day of malaria diagnosis, Immunoglobulin (IgG) antibodies against GLURP, MSP3 and HRP II peptides were present in the blood of 75%, 41% and 60% of patients, respectively. 42 days later, the majority of patients had boosted their serum IgG antibody more than 1.2 fold. The increase in level of IgG antibody against the peptides was not affected by parasite density at diagnosis. The median CD4+ cell counts of PLWHAs and HIV negative individuals were not statistically different, and median post-infection increases in anti-peptide IgG were similar in both groups of patients. Conclusion In the majority (70%) of individuals, an infection of P. falciparum elicits at least 20% increase in level of anti-parasite IgG. This boost in anti-P. falciparum IgG is not affected by parasite density on the day of malaria diagnosis, or by HIV status.
Collapse
Affiliation(s)
- Mark Kaddumukasa
- Department of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | | | - Allan Lugaajju
- College of Health Sciences, Makerere University, Kampala, Uganda
| | - Elly Katabira
- Department of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Kristina E M Persson
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden; Department of Laboratory Medicine, Lund University, Stockholm, Sweden
| | - Mats Wahlgren
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Fred Kironde
- College of Health Sciences, Makerere University, Kampala, Uganda; Habib Medical School, IUIU, Kampala, Uganda
| |
Collapse
|
48
|
Abstract
The development of a highly effective malaria vaccine remains a key goal to aid in the control and eventual eradication of this devastating parasitic disease. The field has made huge strides in recent years, with the first-generation vaccine RTS,S showing modest efficacy in a Phase III clinical trial. The updated 2030 Malaria Vaccine Technology Roadmap calls for a second generation vaccine to achieve 75% efficacy over two years for both Plasmodium falciparum and Plasmodium vivax, and for a vaccine that can prevent malaria transmission. Whole-parasite immunisation approaches and combinations of pre-erythrocytic subunit vaccines are now reporting high-level efficacy, whilst exciting new approaches to the development of blood-stage and transmission-blocking vaccine subunit components are entering clinical development. The development of a highly effective multi-component multi-stage subunit vaccine now appears to be a realistic ambition. This review will cover these recent developments in malaria vaccinology.
Collapse
|
49
|
Conway DJ. Paths to a malaria vaccine illuminated by parasite genomics. Trends Genet 2015; 31:97-107. [PMID: 25620796 PMCID: PMC4359294 DOI: 10.1016/j.tig.2014.12.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/19/2014] [Accepted: 12/19/2014] [Indexed: 11/24/2022]
Abstract
Discovery of vaccine candidate antigens by parasite genome sequence analyses. Genetic crosses, linkage group selection, and functional studies on parasites. Characterizing developmental and epigenetic variation alongside allelic polymorphism. Selection by naturally acquired immune responses helps to focus vaccine design.
More human death and disease is caused by malaria parasites than by all other eukaryotic pathogens combined. As early as the sequencing of the first human genome, malaria parasite genomics was prioritized to fuel the discovery of vaccine candidate antigens. This stimulated increased research on malaria, generating new understanding of the cellular and molecular mechanisms of infection and immunity. This review of recent developments illustrates how new approaches in parasite genomics, and increasingly large amounts of data from population studies, are helping to identify antigens that are promising lead targets. Although these results have been encouraging, effective discovery and characterization need to be coupled with more innovation and funding to translate findings into newly designed vaccine products for clinical trials.
Collapse
Affiliation(s)
- David J Conway
- Pathogen Molecular Biology Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK.
| |
Collapse
|
50
|
Assessment of humoral immune responses to blood-stage malaria antigens following ChAd63-MVA immunization, controlled human malaria infection and natural exposure. PLoS One 2014; 9:e107903. [PMID: 25254500 PMCID: PMC4177865 DOI: 10.1371/journal.pone.0107903] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 08/17/2014] [Indexed: 11/19/2022] Open
Abstract
The development of protective vaccines against many difficult infectious pathogens will necessitate the induction of effective antibody responses. Here we assess humoral immune responses against two antigens from the blood-stage merozoite of the Plasmodium falciparum human malaria parasite--MSP1 and AMA1. These antigens were delivered to healthy malaria-naïve adult volunteers in Phase Ia clinical trials using recombinant replication-deficient viral vectors--ChAd63 to prime the immune response and MVA to boost. In subsequent Phase IIa clinical trials, immunized volunteers underwent controlled human malaria infection (CHMI) with P. falciparum to assess vaccine efficacy, whereby all but one volunteer developed low-density blood-stage parasitemia. Here we assess serum antibody responses against both the MSP1 and AMA1 antigens following i) ChAd63-MVA immunization, ii) immunization and CHMI, and iii) primary malaria exposure in the context of CHMI in unimmunized control volunteers. Responses were also assessed in a cohort of naturally-immune Kenyan adults to provide comparison with those induced by a lifetime of natural malaria exposure. Serum antibody responses against MSP1 and AMA1 were characterized in terms of i) total IgG responses before and after CHMI, ii) responses to allelic variants of MSP1 and AMA1, iii) functional growth inhibitory activity (GIA), iv) IgG avidity, and v) isotype responses (IgG1-4, IgA and IgM). These data provide the first in-depth assessment of the quality of adenovirus-MVA vaccine-induced antibody responses in humans, along with assessment of how these responses are modulated by subsequent low-density parasite exposure. Notable differences were observed in qualitative aspects of the human antibody responses against these malaria antigens depending on the means of their induction and/or exposure of the host to the malaria parasite. Given the continued clinical development of viral vectored vaccines for malaria and a range of other diseases targets, these data should help to guide further immuno-monitoring studies of vaccine-induced human antibody responses.
Collapse
|