1
|
Yun SY, Nguyen MN, Hong H, Bae GJ, Eom TH, Hoang VT, Park H, Yeo SJ. Immunogenicity and efficacy in mice of two adjuvant formulations based on the C -and N-terminus of merozoite surface protein 1 of Plasmodium yoelii. Vaccine 2025; 55:127032. [PMID: 40112557 DOI: 10.1016/j.vaccine.2025.127032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
The carboxyl-terminal fragment of MSP-1 is a potential malaria vaccine candidate, but its limited immunogenicity in humans has slowed clinical progress, needing the optimization of formulation of adjuvant and construct. In this study, the N- and C-terminal fragments of the PyMSP-1 (PyMSP-1 N and PyMSP-1C) were immunized to mice with either incomplete Freund's adjuvant (IFA) plus CpG ODN 1826 or Aluminum salts (Alum) plus CpG, followed by a challenge with Plasmodium yoelii 17XNL to investigate vaccine efficacy. Humoral response and antigen-specific T-cell-derived IFN-γ cytokines were analyzed to compare both fragments. After challenge infection, all mice immunized by PyMSP-1C in IFA plus CpG ODN survived with low-grade parasitemia, while 50 % of mice immunized with PyMSP-1 N in Alum plus CpG ODN died with high levels of parasitemia. Co-immunized with both fragments prevented parasitemia entirely, with IFA plus CpG adjuvants proving more suitable than Alum plus CpG. Both fragments elicited a comparable humoral response when they were formulated with IFA plus CpG ODN but PyMSP-1 N formulated with Alum plus CpG ODN significantly decreased the antigen-specific IgG level. While both IgG1 and IgG2c levels were comparable in two fragments formulated by IFA plus CpG ODN, it was efficient to induce the level of IgG2c of PyMSP-1 N fragment (P < 0.0001). Likewise, IFN-γ from both CD8+ and CD4+ T-cells was significantly lower by PyMSP-1 N than PyMSP-1C formulated in IFA plus CpG ODN (P < 0.0001). In conclusion, the N-terminal fragment of PyMSP-1 protected mice although it showed lower humoral and cellular immune response compared to C-terminal of MSP-1 in IFA plus CpG. The antibody level of PyMSP-1 N was comparable to that of PyMSP-1C when it was formulated with IFA plus CpG.
Collapse
Affiliation(s)
- Su-Yeon Yun
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
| | - Minh-Ngoc Nguyen
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
| | - Hyelee Hong
- Department of Biomedical Sciences, Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Gum-Ju Bae
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
| | - Tae Hui Eom
- Department of Biomedical Sciences, Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Vui Thi Hoang
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
| | - Hyun Park
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
| | - Seon-Ju Yeo
- Department of Biomedical Sciences, Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Tropical Medicine and Parasitology, Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea.
| |
Collapse
|
2
|
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
|
3
|
Hassan I, Kanoi BN, Nagaoka H, Sattabongkot J, Udomsangpetch R, Tsuboi T, Takashima E. High-Throughput Antibody Profiling Identifies Targets of Protective Immunity against P. falciparum Malaria in Thailand. Biomolecules 2023; 13:1267. [PMID: 37627332 PMCID: PMC10452476 DOI: 10.3390/biom13081267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Malaria poses a significant global health challenge, resulting in approximately 600,000 deaths each year. Individuals living in regions with endemic malaria have the potential to develop partial immunity, thanks in part to the presence of anti-plasmodium antibodies. As efforts are made to optimize and implement strategies to reduce malaria transmission and ultimately eliminate the disease, it is crucial to understand how these interventions impact naturally acquired protective immunity. To shed light on this, our study focused on assessing antibody responses to a carefully curated library of P. falciparum recombinant proteins (n = 691) using samples collected from individuals residing in a low-malaria-transmission region of Thailand. We conducted the antibody assays using the AlphaScreen system, a high-throughput homogeneous proximity-based bead assay that detects protein interactions. We observed that out of the 691 variable surface and merozoite stage proteins included in the library, antibodies to 268 antigens significantly correlated with the absence of symptomatic malaria in an univariate analysis. Notably, the most prominent antigens identified were P. falciparum erythrocyte membrane protein 1 (PfEMP1) domains. These results align with our previous research conducted in Uganda, suggesting that similar antigens like PfEMP1s might play a pivotal role in determining infection outcomes in diverse populations. To further our understanding, it remains critical to conduct functional characterization of these identified proteins, exploring their potential as correlates of protection or as targets for vaccine development.
Collapse
Affiliation(s)
- Ifra Hassan
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama 790-8577, Japan; (I.H.); (H.N.)
| | - Bernard N. Kanoi
- Centre for Malaria Elimination, Institute of Tropical Medicine, Mount Kenya University, Thika 01000, Kenya;
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama 790-8577, Japan; (I.H.); (H.N.)
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Rachanee Udomsangpetch
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand;
| | - Takafumi Tsuboi
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama 790-8577, Japan;
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama 790-8577, Japan; (I.H.); (H.N.)
| |
Collapse
|
4
|
Siddiqui AJ, Bhardwaj J, Saxena J, Jahan S, Snoussi M, Bardakci F, Badraoui R, Adnan M. A Critical Review on Human Malaria and Schistosomiasis Vaccines: Current State, Recent Advancements, and Developments. Vaccines (Basel) 2023; 11:vaccines11040792. [PMID: 37112704 PMCID: PMC10146311 DOI: 10.3390/vaccines11040792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/07/2023] Open
Abstract
Malaria and schistosomiasis are two major parasitic diseases that remain leading causes of morbidity and mortality worldwide. Co-infections of these two parasites are common in the tropics, where both diseases are endemic. The clinical consequences of schistosomiasis and malaria are determined by a variety of host, parasitic, and environmental variables. Chronic schistosomiasis causes malnutrition and cognitive impairments in children, while malaria can cause fatal acute infections. There are effective drugs available to treat malaria and schistosomiasis. However, the occurrence of allelic polymorphisms and the rapid selection of parasites with genetic mutations can confer reduced susceptibility and lead to the emergence of drug resistance. Moreover, the successful elimination and complete management of these parasites are difficult due to the lack of effective vaccines against Plasmodium and Schistosoma infections. Therefore, it is important to highlight all current vaccine candidates undergoing clinical trials, such as pre-erythrocytic and erythrocytic stage malaria, as well as a next-generation RTS,S-like vaccine, the R21/Matrix-M vaccine, that conferred 77% protection against clinical malaria in a Phase 2b trial. Moreover, this review also discusses the progress and development of schistosomiasis vaccines. Furthermore, significant information is provided through this review on the effectiveness and progress of schistosomiasis vaccines currently under clinical trials, such as Sh28GST, Sm-14, and Sm-p80. Overall, this review provides insights into recent progress in malarial and schistosomiasis vaccines and their developmental approaches.
Collapse
Affiliation(s)
- Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
| | - Jyoti Bhardwaj
- Division of Infectious Diseases, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Juhi Saxena
- Department of Biotechnology, University Institute of Biotechnology, Chandigarh University, Gharuan, NH-95, Ludhiana—Chandigarh State Hwy, Mohali 140413, India
| | - Sadaf Jahan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia
| | - Mejdi Snoussi
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
- Laboratory of Genetics, Biodiversity and Valorization of Bio-Resources (LR11ES41), Higher Institute of Biotechnology of Monastir, University of Monastir, Avenue TaharHaddas BP74, Monastir 5000, Tunisia
| | - Fevzi Bardakci
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
| | - Riadh Badraoui
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
- Section of Histology-Cytology, Medicine Faculty of Tunis, University of Tunis El Manar, Tunis 1017, Tunisia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
| |
Collapse
|
5
|
Browne DJ, Kelly AM, Brady JL, Doolan DL. A high-throughput screening RT-qPCR assay for quantifying surrogate markers of immunity from PBMCs. Front Immunol 2022; 13:962220. [PMID: 36110843 PMCID: PMC9469018 DOI: 10.3389/fimmu.2022.962220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Immunoassays that quantitate cytokines and other surrogate markers of immunity from peripheral blood mononuclear cells (PBMCs), such as flow cytometry or Enzyme-Linked Immunosorbent Spot (ELIspot), allow highly sensitive measurements of immune effector function. However, those assays consume relatively high numbers of cells and expensive reagents, precluding comprehensive analyses and high-throughput screening (HTS). To address this issue, we developed a sensitive and specific reverse transcription-quantitative PCR (RT-qPCR)-based HTS assay, specifically designed to quantify surrogate markers of immunity from very low numbers of PBMCs. We systematically evaluated the volumes and concentrations of critical reagents within the RT-qPCR protocol, miniaturizing the assay and ultimately reducing the cost by almost 90% compared to current standard practice. We assessed the suitability of this cost-optimized RT-qPCR protocol as an HTS tool and determined the assay exceeds HTS uniformity and signal variance testing standards. Furthermore, we demonstrate this technique can effectively delineate a hierarchy of responses from as little as 50,000 PBMCs stimulated with CD4+ or CD8+ T cell peptide epitopes. Finally, we establish that this HTS-optimized protocol has single-cell analytical sensitivity and a diagnostic sensitivity equivalent to detecting 1:10,000 responding cells (i.e., 100 Spot Forming Cells/106 PBMCs by ELIspot) with over 90% accuracy. We anticipate this assay will have widespread applicability in preclinical and clinical studies, especially when samples are limited, and cost is an important consideration.
Collapse
|
6
|
Takashima E, Kanoi BN, Nagaoka H, Morita M, Hassan I, Palacpac NMQ, Egwang TG, Horii T, Gitaka J, Tsuboi T. Meta-Analysis of Human Antibodies Against Plasmodium falciparum Variable Surface and Merozoite Stage Antigens. Front Immunol 2022; 13:887219. [PMID: 35757771 PMCID: PMC9218060 DOI: 10.3389/fimmu.2022.887219] [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: 03/01/2022] [Accepted: 05/11/2022] [Indexed: 11/30/2022] Open
Abstract
Concerted efforts to fight malaria have caused significant reductions in global malaria cases and mortality. Sustaining this will be critical to avoid rebound and outbreaks of seasonal malaria. Identifying predictive attributes that define clinical malaria will be key to guide development of second-generation tools to fight malaria. Broadly reactive antibodies against variable surface antigens that are expressed on the surface of infected erythrocytes and merozoites stage antigens are targets of naturally acquired immunity and prime candidates for anti-malaria therapeutics and vaccines. However, predicting the relationship between the antigen-specific antibodies and protection from clinical malaria remains unresolved. Here, we used new datasets and multiple approaches combined with re-analysis of our previous data to assess the multi-dimensional and complex relationship between antibody responses and clinical malaria outcomes. We observed 22 antigens (17 PfEMP1 domains, 3 RIFIN family members, merozoite surface protein 3 (PF3D7_1035400), and merozoites-associated armadillo repeats protein (PF3D7_1035900) that were selected across three different clinical malaria definitions (1,000/2,500/5,000 parasites/µl plus fever). In addition, Principal Components Analysis (PCA) indicated that the first three components (Dim1, Dim2 and Dim3 with eigenvalues of 306, 48, and 29, respectively) accounted for 66.1% of the total variations seen. Specifically, the Dim1, Dim2 and Dim3 explained 52.8%, 8.2% and 5% of variability, respectively. We further observed a significant relationship between the first component scores and age with antibodies to PfEMP1 domains being the key contributing variables. This is consistent with a recent proposal suggesting that there is an ordered acquisition of antibodies targeting PfEMP1 proteins. Thus, although limited, and further work on the significance of the selected antigens will be required, these approaches may provide insights for identification of drivers of naturally acquired protective immunity as well as guide development of additional tools for malaria elimination and eradication.
Collapse
Affiliation(s)
- Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Bernard N Kanoi
- Centre for Research in Infectious Diseases, Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Masayuki Morita
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Ifra Hassan
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Nirianne M Q Palacpac
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | | | - Toshihiro Horii
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Jesse Gitaka
- Centre for Research in Infectious Diseases, Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
| | - Takafumi Tsuboi
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama, Japan
| |
Collapse
|
7
|
Goodswen SJ, Kennedy PJ, Ellis JT. Compilation of parasitic immunogenic proteins from 30 years of published research using machine learning and natural language processing. Sci Rep 2022; 12:10349. [PMID: 35725870 PMCID: PMC9208253 DOI: 10.1038/s41598-022-13790-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 05/18/2022] [Indexed: 12/02/2022] Open
Abstract
The World Health Organisation reported in 2020 that six of the top 10 sources of death in low-income countries are parasites. Parasites are microorganisms in a relationship with a larger organism, the host. They acquire all benefits at the host’s expense. A disease develops if the parasitic infection disrupts normal functioning of the host. This disruption can range from mild to severe, including death. Humans and livestock continue to be challenged by established and emerging infectious disease threats. Vaccination is the most efficient tool for preventing current and future threats. Immunogenic proteins sourced from the disease-causing parasite are worthwhile vaccine components (subunits) due to reliable safety and manufacturing capacity. Publications with ‘subunit vaccine’ in their title have accumulated to thousands over the last three decades. However, there are possibly thousands more reporting immunogenicity results without mentioning ‘subunit’ and/or ‘vaccine’. The exact number is unclear given the non-standardised keywords in publications. The study aim is to identify parasite proteins that induce a protective response in an animal model as reported in the scientific literature within the last 30 years using machine learning and natural language processing. Source code to fulfil this aim and the vaccine candidate list obtained is made available.
Collapse
Affiliation(s)
- Stephen J Goodswen
- School of Life Sciences, University of Technology Sydney, 15 Broadway, Ultimo, NSW, 2007, Australia
| | - Paul J Kennedy
- School of Computer Science, Faculty of Engineering and Information Technology and the Australian Artificial Intelligence Institute, University of Technology Sydney, 15 Broadway, Ultimo, NSW, 2007, Australia
| | - John T Ellis
- School of Life Sciences, University of Technology Sydney, 15 Broadway, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
8
|
Zhang X, Wei H, Zhang Y, Zhao Y, Wang L, Hu Y, Nguitragool W, Sattabongkot J, Adams J, Cui L, Cao Y, Wang Q. Genetic diversity of Plasmodium vivax reticulocyte binding protein 2b in global parasite populations. Parasit Vectors 2022; 15:205. [PMID: 35698238 PMCID: PMC9191549 DOI: 10.1186/s13071-022-05296-6] [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: 10/27/2021] [Accepted: 04/23/2022] [Indexed: 11/19/2022] Open
Abstract
Background Plasmodium vivax reticulocyte binding protein 2b (PvRBP2b) plays a critical role in parasite invasion of reticulocytes by binding the transferrin receptor 1. PvRBP2b is a vaccine candidate based on the negative correlation between antibody titers against PvRBP2b recombinant proteins and parasitemia and risk of vivax malaria. The aim of this study was to analyze the genetic diversity of the PvRBP2b gene in the global P. vivax populations. Methods Near full-length PvRBP2b nucleotide sequences (190–8349 bp) were obtained from 88 P. vivax isolates collected from the China–Myanmar border (n = 44) and Thailand (n = 44). An additional 224 PvRBP2b sequences were retrieved from genome sequences from parasite populations worldwide. The genetic diversity, neutral selection, haplotype distribution and genetic differentiation of PvRBP2b were examined. Results The genetic diversity of PvRBP2b was distributed unevenly, with peak diversity found in the reticulocyte binding region in the N-terminus. Neutrality analysis suggested that this region is subjected to balancing selection or population bottlenecks. Several amino acid variants were found in all or nearly all P. vivax endemic regions. However, the critical residues responsible for reticulocyte binding were highly conserved. There was substantial population differentiation according to the geographical separation. The distribution of haplotypes in the reticulocyte binding region varied among regions; even the two major haplotypes Hap_6 and Hap_8 were found in only five populations. Conclusions Our data show considerable genetic variations of PvRBPb in global parasite populations. The geographic divergence may pose a challenge to PvRBP2b-based vaccine development. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-022-05296-6.
Collapse
Affiliation(s)
- Xuexing Zhang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China
| | - Haichao Wei
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China.,Department of Blood Transfusion Medicine, General Hospital of Northern Theater Command, Shenyang, 110015, Liaoning, China
| | - Yangminghui Zhang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yan Zhao
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China
| | - Lin Wang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China.,Department of Blood Transfusion, Yantaishan Hospital, Yantai, 264000, Shandong, China
| | - Yubing Hu
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China.,Central Laboratory, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Wang Nguitragool
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - John Adams
- College of Public Health, Global Health Infectious Disease Research (GHIDR) Program, Tampa, FL, USA
| | - Liwang Cui
- College of Public Health, Global Health Infectious Disease Research (GHIDR) Program, Tampa, FL, USA.,Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Suite 304, Tampa, FL, 33612, USA
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Qinghui Wang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
9
|
Takashima E, Tachibana M, Morita M, Nagaoka H, Kanoi BN, Tsuboi T. Identification of Novel Malaria Transmission-Blocking Vaccine Candidates. Front Cell Infect Microbiol 2021; 11:805482. [PMID: 34917521 PMCID: PMC8670312 DOI: 10.3389/fcimb.2021.805482] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 11/16/2021] [Indexed: 01/02/2023] Open
Abstract
Control measures have significantly reduced malaria morbidity and mortality in the last two decades; however, the downward trends have stalled and have become complicated by the emergence of COVID-19. Significant efforts have been made to develop malaria vaccines, but currently only the RTS,S/AS01 vaccine against Plasmodium falciparum has been recommended by the WHO, for widespread use among children in sub-Saharan Africa. The efficacy of RTS,S/AS01 is modest, and therefore the development of more efficacious vaccines is still needed. In addition, the development of transmission-blocking vaccines (TBVs) to reduce the parasite transmission from humans to mosquitoes is required toward the goal of malaria elimination. Few TBVs have reached clinical development, and challenges include low immunogenicity or high reactogenicity in humans. Therefore, novel approaches to accelerate TBV research and development are urgently needed, especially novel TBV candidate discovery. In this mini review we summarize the progress in TBV research and development, novel TBV candidate discovery, and discuss how to accelerate novel TBV candidate discovery.
Collapse
Affiliation(s)
- Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Mayumi Tachibana
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Japan
| | - Masayuki Morita
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Bernard N Kanoi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Takafumi Tsuboi
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama, Japan
| |
Collapse
|
10
|
Kanoi BN, Nagaoka H, White MT, Morita M, Palacpac NMQ, Ntege EH, Balikagala B, Yeka A, Egwang TG, Horii T, Tsuboi T, Takashima E. Global Repertoire of Human Antibodies Against Plasmodium falciparum RIFINs, SURFINs, and STEVORs in a Malaria Exposed Population. Front Immunol 2020; 11:893. [PMID: 32477363 PMCID: PMC7235171 DOI: 10.3389/fimmu.2020.00893] [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: 01/10/2020] [Accepted: 04/17/2020] [Indexed: 11/15/2022] Open
Abstract
Clinical immunity to malaria develops after repeated exposure to Plasmodium falciparum parasites. Broadly reactive antibodies against parasite antigens expressed on the surface of infected erythrocytes (variable surface antigens; VSAs) are candidates for anti-malaria therapeutics and vaccines. Among the VSAs, several RIFIN, STEVOR, and SURFIN family members have been demonstrated to be targets of naturally acquired immunity against malaria. For example, RIFIN family members are important ligands for opsonization of P. falciparum infected erythrocytes with specific immunoglobulins (IgG) acquiring broad protective reactivity. However, the global repertoire of human anti-VSAs IgG, its variation in children, and the key protective targets remain poorly understood. Here, we report wheat germ cell-free system-based production and serological profiling of a comprehensive library of A-RIFINs, B-RIFINs, STEVORs, and SURFINs derived from the P. falciparum 3D7 parasite strain. We observed that >98% of assayed proteins (n = 265) were immunogenic in malaria-exposed individuals in Uganda. The overall breadth of immune responses was significantly correlated with age but not with clinical malaria outcome among the study volunteers. However, children with high levels of antibodies to four RIFINs (PF3D7_0201000, PF3D7_1254500, PF3D7_1040600, PF3D7_1041100), STEVOR (PF3D7_0732000), and SURFIN 1.2 (PF3D7_0113600) had prospectively reduced the risk of developing febrile malaria, suggesting that the 5 antigens are important targets of protective immunity. Further studies on the significance of repeated exposure to malaria infection and maintenance of such high-level antibodies would contribute to a better understanding of susceptibility and naturally acquired immunity to malaria.
Collapse
Affiliation(s)
- Bernard N Kanoi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Michael T White
- Department of Parasites and Insect Vectors, Pasteur Institute, Paris, France
| | - Masayuki Morita
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Nirianne M Q Palacpac
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Edward H Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine and Hospital, University of the Ryukyus, Okinawa, Japan
| | - Betty Balikagala
- Department of Tropical Medicine and Parasitology, School of Medicine, Juntendo University, Tokyo, Japan
| | - Adoke Yeka
- Makerere University School of Public Health, Kampala, Uganda
| | | | - Toshihiro Horii
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| |
Collapse
|
11
|
Longley RJ, White MT, Takashima E, Brewster J, Morita M, Harbers M, Obadia T, Robinson LJ, Matsuura F, Liu ZSJ, Li-Wai-Suen CSN, Tham WH, Healer J, Huon C, Chitnis CE, Nguitragool W, Monteiro W, Proietti C, Doolan DL, Siqueira AM, Ding XC, Gonzalez IJ, Kazura J, Lacerda M, Sattabongkot J, Tsuboi T, Mueller I. Development and validation of serological markers for detecting recent Plasmodium vivax infection. Nat Med 2020; 26:741-749. [PMID: 32405064 DOI: 10.1038/s41591-020-0841-4] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 03/18/2020] [Indexed: 11/09/2022]
Abstract
A major gap in the Plasmodium vivax elimination toolkit is the identification of individuals carrying clinically silent and undetectable liver-stage parasites, called hypnozoites. This study developed a panel of serological exposure markers capable of classifying individuals with recent P. vivax infections who have a high likelihood of harboring hypnozoites. We measured IgG antibody responses to 342 P. vivax proteins in longitudinal clinical cohorts conducted in Thailand and Brazil and identified candidate serological markers of exposure. Candidate markers were validated using samples from year-long observational cohorts conducted in Thailand, Brazil and the Solomon Islands and antibody responses to eight P. vivax proteins classified P. vivax infections in the previous 9 months with 80% sensitivity and specificity. Mathematical models demonstrate that a serological testing and treatment strategy could reduce P. vivax prevalence by 59-69%. These eight antibody responses can serve as a biomarker, identifying individuals who should be targeted with anti-hypnozoite therapy.
Collapse
Affiliation(s)
- Rhea J Longley
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Michael T White
- Unité Malaria: Parasites et Hôtes, Département Parasites et Insectes Vecteurs, Institut Pasteur, Paris, France
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Jessica Brewster
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Masayuki Morita
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Matthias Harbers
- CellFree Sciences Co., Ltd., Yokohama, Japan
- RIKEN Center for Integrated Medical Sciences (IMS), Yokohama, Japan
| | - Thomas Obadia
- Unité Malaria: Parasites et Hôtes, Département Parasites et Insectes Vecteurs, Institut Pasteur, Paris, France
- Hub de Bioinformatique et Biostatistique, Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, Paris, France
| | - Leanne J Robinson
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
- Burnet Institute, Melbourne, Victoria, Australia
| | | | - Zoe S J Liu
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Connie S N Li-Wai-Suen
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Wai-Hong Tham
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Julie Healer
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Christele Huon
- Malaria Parasite Biology and Vaccines, Department of Parasites & Insect Vectors, Institut Pasteur, Paris, France
| | - Chetan E Chitnis
- Malaria Parasite Biology and Vaccines, Department of Parasites & Insect Vectors, Institut Pasteur, Paris, France
| | - Wang Nguitragool
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wuelton Monteiro
- Fundacão de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
- Universidade do Estado do Amazonas, Manaus, Brazil
| | - Carla Proietti
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Denise L Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Andre M Siqueira
- Instituto Nacional de Infectologia Evandro Chagas-Fiocruz, Rio de Janeiro, Brazil
| | - Xavier C Ding
- Foundation for Innovative New Diagnostics, Geneva, Switzerland
| | | | - James Kazura
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, USA
| | - Marcus Lacerda
- Fundacão de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
- Instituto Leônidas & Maria Deane (Fiocruz), Manaus, Brazil
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Ivo Mueller
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia.
- Unité Malaria: Parasites et Hôtes, Département Parasites et Insectes Vecteurs, Institut Pasteur, Paris, France.
| |
Collapse
|
12
|
Lee SK, Han JH, Park JH, Ha KS, Park WS, Hong SH, Na S, Cheng Y, Han ET. Evaluation of antibody responses to the early transcribed membrane protein family in Plasmodium vivax. Parasit Vectors 2019; 12:594. [PMID: 31856917 PMCID: PMC6921578 DOI: 10.1186/s13071-019-3846-4] [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: 09/25/2019] [Accepted: 12/09/2019] [Indexed: 11/25/2022] Open
Abstract
Background Malaria parasites form intracellular membranes that separate the parasite from the internal space of erythrocytes, and membrane proteins from the parasites are exported to the host via the membrane. In our previous study, Plasmodium vivax early transcribed membrane protein (PvETRAMP) 11.2, an intracellular membrane protein that is highly expressed in blood-stage parasites, was characterized as a highly immunogenic protein in P. vivax malaria patients. However, the other PvETRAMP family proteins have not yet been investigated. In this study, PvETRAMPs were expressed and evaluated to determine their immunological profiles. Methods The protein structure and amino acid alignment were carried out using bioinformatics analysis software. A total of six PvETRAMP family proteins were successfully expressed and purified using a wheat germ cell free protein expression system and the purified proteins were used for protein microarray and immunization of mice. The localization of the protein was determined with serum against PvETRAMP4. IgG subclasses were assessed from the immunized mice. Results In silico analysis showed that P. vivax exhibits nine genes encoding the ETRAMP family. The ETRAMP family proteins are relatively small molecules with conserved structural features. A total of 6 recombinant ETRAMP proteins were successfully expressed and purified. The serum positivity of P. vivax malaria patients and healthy individuals was evaluated using a protein microarray method. Among the PvETRAMPs, ETRAMP4 showed the highest positivity rate of 62%, comparable to that of PvETRAMP11.2, which served as the positive control, and a typical export pattern of PvETRAMP4 was observed in the P. vivax parasite. The assessment of IgG subclasses in mice immunized with PvETRAMP4 showed high levels of IgG1 and IgG2b. PvETRAMP family proteins were identified and characterized as serological markers. Conclusions The relatively high antibody responses to PvETRAMP4 as well as the specific IgG subclasses observed in immunized mice suggest that the ETRAMP family is immunogenic in pathogens and can be used as a protein marker and for vaccine development.![]()
Collapse
Affiliation(s)
- Seong-Kyun Lee
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Ji-Hoon Park
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Kwon-Soo Ha
- Department of Cellular and Molecular Biology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Won Sun Park
- Department of Physiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Sunghun Na
- Department of Obstetrics and Gynecology, Kangwon National University Hospital, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Yang Cheng
- Department of Public Health and Preventive Medicine, Laboratory of Pathogen Infection and Immunity, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, People's Republic of China.
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea.
| |
Collapse
|
13
|
Tran TM, Crompton PD. Decoding the complexities of human malaria through systems immunology. Immunol Rev 2019; 293:144-162. [PMID: 31680289 DOI: 10.1111/imr.12817] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/18/2022]
Abstract
The complexity of the Plasmodium parasite and its life cycle poses a challenge to our understanding of the host immune response against malaria. Studying human immune responses during natural and experimental Plasmodium infections can enhance our understanding of malaria-protective immunity and inform the design of disease-modifying adjunctive therapies and next-generation malaria vaccines. Systems immunology can complement conventional approaches to facilitate our understanding of the complex immune response to the highly dynamic malaria parasite. In this review, recent studies that used systems-based approaches to evaluate human immune responses during natural and experimental Plasmodium falciparum and Plasmodium vivax infections as well as during immunization with candidate malaria vaccines are summarized and related to each other. The potential for next-generation technologies to address the current limitations of systems-based studies of human malaria are discussed.
Collapse
Affiliation(s)
- Tuan M Tran
- Division of Infectious Diseases, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Ryan White Center for Pediatric Infectious Disease and Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Peter D Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
14
|
Miura K, Tachibana M, Takashima E, Morita M, Kanoi BN, Nagaoka H, Baba M, Torii M, Ishino T, Tsuboi T. Malaria transmission-blocking vaccines: wheat germ cell-free technology can accelerate vaccine development. Expert Rev Vaccines 2019; 18:1017-1027. [PMID: 31566026 PMCID: PMC11000147 DOI: 10.1080/14760584.2019.1674145] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 09/25/2019] [Indexed: 12/18/2022]
Abstract
Introduction: Highly effective malaria vaccines are essential component toward malaria elimination. Although the leading malaria vaccine, RTS,S/AS01, with modest efficacy is being evaluated in a pilot feasibility trial, development of a malaria transmission-blocking vaccine (TBV) could make a major contribution toward malaria elimination. Only a few TBV antigens have reached pre-clinical or clinical development but with several challenges including difficulties in the expression of malaria recombinant proteins and low immunogenicity in humans. Therefore, novel approaches to accelerate TBV research to preclinical development are critical to generate an efficacious TBV.Areas covered: PubMed was searched to review the progress and future prospects of malaria TBV research and development. We also reviewed registered trials at ClinicalTrials.gov as well as post-genome TBV candidate discovery research including our efforts.Expert opinion: Wheat germ cell-free protein synthesis technology can accelerate TBV development by overcoming some current challenges of TBV research.
Collapse
Affiliation(s)
- Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Mayumi Tachibana
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Japan
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Masayuki Morita
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Bernard N Kanoi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Minami Baba
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Japan
| | - Motomi Torii
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Japan
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Tomoko Ishino
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| |
Collapse
|
15
|
Identification of domains within Pfs230 that elicit transmission blocking antibody responses. Vaccine 2019; 37:1799-1806. [PMID: 30824357 PMCID: PMC6708081 DOI: 10.1016/j.vaccine.2019.02.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/23/2019] [Accepted: 02/05/2019] [Indexed: 11/23/2022]
Abstract
A transmission-blocking vaccine (TBV) against Plasmodium falciparum is likely to be a valuable tool in a malaria eradication program. Pfs230 is one of the major TBV candidates, and multiple Pfs230-based vaccines induced antibodies, which prevented oocyst formation in mosquitoes as determined by a standard membrane-feeding assay (SMFA). Pfs230 is a >300 kDa protein consisting of 14 cysteine motif (CM) domains, and the size and cysteine-rich nature of the molecule have hampered its production as an intact protein. Except for one early study with maltose-binding protein fusion Pfs230 constructs expressed in Esherichia coli, all other studies have focused on only the first four CM domains in the Pfs230 molecule. To identify all possible TBV candidate domains, we systematically produced either single-CM-domain (a total of 14), 2-CM-domain (7), or 4-CM-domain (6) recombinant protein fragments using a eukaryotic wheat germ cell-free expression system (WGCFS). In addition, two more constructs which covered previously published regions, and an N-terminal prodomain construct spanning the natural cleavage site of Pfs230 were produced. Antisera against each fragment were generated in mice and we evaluated the reactivity to native Pfs230 protein by Western blots and immunofluorescence assay (IFA), and functionality by SMFA. All 30 WGCFS-produced Pfs230 constructs were immunogenic in mice. Approximately half of the mouse antibodies specifically recognized native Pfs230 by Western blots with variable band intensities. Among them, seven antibodies showed higher reactivities against native Pfs230 determined by IFA. Interestingly, antibodies against all protein fragments containing CM domain 1 displayed strong inhibitions in SMFA, while antibodies generated using constructs without CM domain 1 showed no inhibition. The results strongly support the concept that future Pfs230-based vaccine development should focus on the Pfs230 CM domain 1.
Collapse
|
16
|
Ito D, Takashima E, Yamasaki T, Hatano S, Hasegawa T, Miura K, Morita M, Thongkukiatkul A, Diakite M, Long CA, Sattabongkot J, Udomsangpetch R, Iriko H, Ishino T, Tsuboi T. Antibodies against a Plasmodium falciparum RON12 inhibit merozoite invasion into erythrocytes. Parasitol Int 2018; 68:87-91. [PMID: 30342119 DOI: 10.1016/j.parint.2018.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/10/2018] [Accepted: 10/16/2018] [Indexed: 10/28/2022]
Abstract
Proteins coating Plasmodium merozoite surface and secreted from its apical organelles are considered as promising vaccine candidates for blood-stage malaria. The rhoptry neck protein 12 of Plasmodium falciparum (PfRON12) was recently reported as a protein specifically expressed in schizonts and localized to the rhoptry neck of merozoites. Here, we assessed its potential as a vaccine candidate. We expressed a recombinant PfRON12 protein by a wheat germ cell-free system to obtain anti-PfRON12 antibody. Immunoblot analysis of schizont lysates detected a single band at approximately 40 kDa under reducing conditions, consistent with the predicted molecular weight. Additionally, anti-PfRON12 antibody recognized a single band around 80 kDa under non-reducing conditions, suggesting native PfRON12 forms a disulfide-bond-mediated multimer. Immunofluorescence assay and immunoelectron microscopy revealed that PfRON12 localized to the rhoptry neck of merozoites in schizonts and to the surface of free merozoites. The biological activity of anti-PfRON12 antibody was tested by in vitro growth inhibition assay (GIA), and the rabbit antibodies significantly inhibited merozoite invasion of erythrocytes. We then investigated whether PfRON12 is immunogenic in P. falciparum-infected individuals. The sera from P. falciparum infected individuals in Thailand and Mali reacted with the recombinant PfRON12. Furthermore, human anti-PfRON12 antibodies affinity-purified from Malian serum samples inhibited merozoite invasion of erythrocytes in vitro. Moreover, pfron12 is highly conserved with only 4 non-synonymous mutations in the coding sequence from approximately 200 isolates deposited in PlasmoDB. These results suggest that PfRON12 might be a potential blood-stage vaccine candidate antigen against P. falciparum.
Collapse
Affiliation(s)
- Daisuke Ito
- Division of Medical Zoology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan.
| | - Tsutomu Yamasaki
- Department of Molecular and Clinical Diagnosis, School of Pharmacy, Shujitsu University, Okayama 703-8516, Japan
| | - Shinya Hatano
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Tomoyuki Hasegawa
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, United States
| | - Masayuki Morita
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Amporn Thongkukiatkul
- Department of Biology, Faculty of Science, Burapha University, Chonburi 20131, Thailand
| | - Mahamadou Diakite
- Malaria Research and Training Centre, Faculty of Medicine, Pharmacy and Odonto-stomatology, University of Sciences, Techniques, and Technologies of Bamako, Point G, BP 1805, Mali
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, United States
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Rachanee Udomsangpetch
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakhosn Pathom, 73170, Thailand
| | - Hideyuki Iriko
- Division of Global Infectious Diseases, Department of Public Health, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo 654-0142, Japan
| | - Tomoko Ishino
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon Ehime, 791-0295, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan.
| |
Collapse
|
17
|
Comprehensive analysis of antibody responses to Plasmodium falciparum erythrocyte membrane protein 1 domains. Vaccine 2018; 36:6826-6833. [PMID: 30262245 DOI: 10.1016/j.vaccine.2018.08.058] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/03/2018] [Accepted: 08/22/2018] [Indexed: 12/31/2022]
Abstract
Acquired antibodies directed towards antigens expressed on the surface of merozoites and infected erythrocytes play an important role in protective immunity to Plasmodium falciparum malaria. P. falciparum erythrocyte membrane protein 1 (PfEMP1), the major parasite component of the infected erythrocyte surface, has been implicated in malaria pathology, parasite sequestration and host immune evasion. However, the extent to which unique PfEMP1 domains interact with host immune response remains largely unknown. In this study, we sought to comprehensively understand the naturally acquired antibody responses targeting different Duffy binding-like (DBL), and Cysteine-rich interdomain region (CIDR) domains in a Ugandan cohort. Consequently, we created a protein library consisting of full-length DBL (n = 163) and CIDR (n = 108) domains derived from 62-var genes based on 3D7 genome. The proteins were expressed by a wheat germ cell-free system; a system that yields plasmodial proteins that are comparatively soluble, intact, biologically active and immunoreactive to human sera. Our findings suggest that all PfEMP1 DBL and CIDR domains, regardless of PfEMP1 group, are targets of naturally acquired immunity. The breadth of the immune response expands with children's age. We concurrently identified 10 DBL and 8 CIDR domains whose antibody responses were associated with reduced risk to symptomatic malaria in the Ugandan children cohort. This study highlights that only a restricted set of specific domains are essential for eliciting naturally acquired protective immunity in malaria. In light of current data, tandem domains in PfEMP1s PF3D7_0700100 and PF3D7_0425800 (DC4) are recommended for extensive evaluation in larger population cohorts to further assess their potential as alternative targets for malaria vaccine development.
Collapse
|
18
|
Othman AS, Lin JW, Franke-Fayard BM, Kroeze H, van Pul FJA, Chevalley-Maurel S, Ramesar J, Marin-Mogollon C, Jore MM, Morin MJ, Long CA, Sauerwein R, Birkett A, Miura K, Janse CJ, Khan SM. Expression of full-length Plasmodium falciparum P48/45 in P. berghei blood stages: A method to express and evaluate vaccine antigens. Mol Biochem Parasitol 2018; 224:44-49. [PMID: 30053393 PMCID: PMC11040549 DOI: 10.1016/j.molbiopara.2018.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/10/2018] [Accepted: 07/19/2018] [Indexed: 12/16/2022]
Abstract
The transmission-blocking vaccine candidate Pfs48/45 from the human malaria parasite Plasmodium falciparum is known to be difficult to express in heterologous systems, either as full-length protein or as correctly folded protein fragments that retain conformational epitopes. In this study we express full-length Pfs48/45 in the rodent parasite P. berghei. Pfs48/45 is expressed as a transgene under control of the strong P. berghei schizont-specific msp1 gene promoter (Pfs48/45@PbMSP1). Pfs48/45@PbMSP1 schizont-infected red blood cells produced full-length Pfs48/45 and the structural integrity of Pfs48/45 was confirmed using a panel of conformation-specific monoclonal antibodies that bind to different Pfs48/45 epitopes. Sera from mice immunized with transgenic Pfs48/45@PbMSP1 schizonts showed strong transmission-reducing activity in mosquitoes infected with P. falciparum using standard membrane feeding. These results demonstrate that transgenic rodent malaria parasites expressing human malaria antigens may be used as means to evaluate immunogenicity and functionality of difficult to express malaria vaccine candidate antigens.
Collapse
Affiliation(s)
- Ahmad Syibli Othman
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, The Netherlands; Faculty of Health Sciences, Universiti Sultan Zainal Abidin, Terengganu, Malaysia
| | - Jing-Wen Lin
- Division of Pediatric Infectious Diseases, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaboration Innovation Center, Chengdu, China
| | - Blandine M Franke-Fayard
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Hans Kroeze
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Fiona J A van Pul
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Séverine Chevalley-Maurel
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Jai Ramesar
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Catherin Marin-Mogollon
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Matthijs M Jore
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, Maryland, USA
| | - Robert Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, Maryland, USA
| | - Chris J Janse
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Shahid M Khan
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, The Netherlands.
| |
Collapse
|