1
|
Safety, immunogenicity and efficacy of PfSPZ Vaccine against malaria in infants in western Kenya: a double-blind, randomized, placebo-controlled phase 2 trial. Nat Med 2021; 27:1636-1645. [PMID: 34518679 DOI: 10.1038/s41591-021-01470-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 07/16/2021] [Indexed: 01/13/2023]
Abstract
The radiation-attenuated Plasmodium falciparum sporozoite (PfSPZ) vaccine provides protection against P. falciparum infection in malaria-naïve adults. Preclinical studies show that T cell-mediated immunity is required for protection and is readily induced in humans after vaccination. However, previous malaria exposure can limit immune responses and vaccine efficacy (VE) in adults. We hypothesized that infants with less previous exposure to malaria would have improved immunity and protection. We conducted a multi-arm, randomized, double-blind, placebo-controlled trial in 336 infants aged 5-12 months to determine the safety, tolerability, immunogenicity and efficacy of the PfSPZ Vaccine in infants in a high-transmission malaria setting in western Kenya ( NCT02687373 ). Groups of 84 infants each received 4.5 × 105, 9.0 × 105 or 1.8 × 106 PfSPZ Vaccine or saline three times at 8-week intervals. The vaccine was well tolerated; 52 (20.6%) children in the vaccine groups and 20 (23.8%) in the placebo group experienced related solicited adverse events (AEs) within 28 d postvaccination and most were mild. There was 1 grade 3-related solicited AE in the vaccine group (0.4%) and 2 in the placebo group (2.4%). Seizures were more common in the highest-dose group (14.3%) compared to 6.0% of controls, with most being attributed to malaria. There was no significant protection against P. falciparum infection in any dose group at 6 months (VE in the 9.0 × 105 dose group = -6.5%, P = 0.598, the primary statistical end point of the study). VE against clinical malaria 3 months after the last dose in the highest-dose group was 45.8% (P = 0.027), an exploratory end point. There was a dose-dependent increase in antibody responses that correlated with VE at 6 months in the lowest- and highest-dose groups. T cell responses were undetectable across all dose groups. Detection of Vδ2+Vγ9+ T cells, which have been correlated with induction of PfSPZ Vaccine T cell immunity and protection in adults, were infrequent. These data suggest that PfSPZ Vaccine-induced T cell immunity is age-dependent and may be influenced by Vδ2+Vγ9+ T cell frequency. Since there was no significant VE at 6 months in these infants, these vaccine regimens will likely not be pursued further in this age group.
Collapse
|
2
|
Reuling IJ, Mendes AM, de Jong GM, Fabra-García A, Nunes-Cabaço H, van Gemert GJ, Graumans W, Coffeng LE, de Vlas SJ, Yang ASP, Lee C, Wu Y, Birkett AJ, Ockenhouse CF, Koelewijn R, van Hellemond JJ, van Genderen PJJ, Sauerwein RW, Prudêncio M. An open-label phase 1/2a trial of a genetically modified rodent malaria parasite for immunization against Plasmodium falciparum malaria. Sci Transl Med 2021; 12:12/544/eaay2578. [PMID: 32434846 DOI: 10.1126/scitranslmed.aay2578] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 04/22/2020] [Indexed: 12/14/2022]
Abstract
For some diseases, successful vaccines have been developed using a nonpathogenic counterpart of the causative microorganism of choice. The nonpathogenicity of the rodent Plasmodium berghei (Pb) parasite in humans prompted us to evaluate its potential as a platform for vaccination against human infection by Plasmodium falciparum (Pf), a causative agent of malaria. We hypothesized that the genetic insertion of a leading protein target for clinical development of a malaria vaccine, Pf circumsporozoite protein (CSP), in its natural pre-erythrocytic environment, would enhance Pb's capacity to induce protective immunity against Pf infection. Hence, we recently generated a transgenic Pb sporozoite immunization platform expressing PfCSP (PbVac), and we now report the clinical evaluation of its biological activity against controlled human malaria infection (CHMI). This first-in-human trial shows that PbVac is safe and well tolerated, when administered by a total of ~300 PbVac-infected mosquitoes per volunteer. Although protective efficacy evaluated by CHMI showed no sterile protection at the tested dose, significant delays in patency (2.2 days, P = 0.03) and decreased parasite density were observed after immunization, corresponding to an estimated 95% reduction in Pf liver parasite burden (confidence interval, 56 to 99%; P = 0.010). PbVac elicits dose-dependent cross-species cellular immune responses and functional PfCSP-dependent antibody responses that efficiently block Pf sporozoite invasion of liver cells in vitro. This study demonstrates that PbVac immunization elicits a marked biological effect, inhibiting a subsequent infection by the human Pf parasite, and establishes the clinical validation of a new paradigm in malaria vaccination.
Collapse
Affiliation(s)
- Isaie J Reuling
- Radboud Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - António M Mendes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Gerdie M de Jong
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, 3015 GD Rotterdam, Netherlands
| | - Amanda Fabra-García
- Radboud Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Helena Nunes-Cabaço
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Geert-Jan van Gemert
- Radboud Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Wouter Graumans
- Radboud Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Luc E Coffeng
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, Netherlands
| | - Sake J de Vlas
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, Netherlands
| | - Annie S P Yang
- Radboud Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Cynthia Lee
- PATH's Malaria Vaccine Initiative, Washington, DC 20001, USA
| | - Yimin Wu
- PATH's Malaria Vaccine Initiative, Washington, DC 20001, USA
| | | | | | - Rob Koelewijn
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, 3015 GD Rotterdam, Netherlands
| | - Jaap J van Hellemond
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, 3015 GD Rotterdam, Netherlands
| | - Perry J J van Genderen
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, 3015 GD Rotterdam, Netherlands. .,Corporate Travel Clinic Erasmus MC, 3015 CP Rotterdam, Netherlands
| | - Robert W Sauerwein
- Radboud Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands.
| | - Miguel Prudêncio
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal.
| |
Collapse
|
3
|
Identification of a neutralizing epitope within minor repeat region of Plasmodium falciparum CS protein. NPJ Vaccines 2021; 6:10. [PMID: 33462218 PMCID: PMC7813878 DOI: 10.1038/s41541-020-00272-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 12/14/2020] [Indexed: 11/08/2022] Open
Abstract
Malaria remains a major cause of morbidity and mortality worldwide with 219 million infections and 435,000 deaths predominantly in Africa. The infective Plasmodium sporozoite is the target of a potent humoral immune response that can protect murine, simian and human hosts against challenge by malaria-infected mosquitoes. Early murine studies demonstrated that sporozoites or subunit vaccines based on the sporozoite major surface antigen, the circumsporozoite (CS) protein, elicit antibodies that primarily target the central repeat region of the CS protein. In the current murine studies, using monoclonal antibodies and polyclonal sera obtained following immunization with P. falciparum sporozoites or synthetic repeat peptides, we demonstrate differences in the ability of these antibodies to recognize the major and minor repeats contained in the central repeat region. The biological relevance of these differences in fine specificity was explored using a transgenic P. berghei rodent parasite expressing the P. falciparum CS repeat region. In these in vitro and in vivo studies, we demonstrate that the minor repeat region, comprised of three copies of alternating NANP and NVDP tetramer repeats, contains an epitope recognized by sporozoite-neutralizing antibodies. In contrast, murine monoclonal antibodies specific for the major CS repeats (NANP)n could be isolated from peptide-immunized mice that had limited or no sporozoite-neutralizing activity. These studies highlight the importance of assessing the fine specificity and functions of antirepeat antibodies elicited by P. falciparum CS-based vaccines and suggest that the design of immunogens to increase antibody responses to minor CS repeats may enhance vaccine efficacy.
Collapse
|
4
|
Protective efficacy of peptides from Plasmodium vivax circumsporozoite protein. Vaccine 2020; 38:4346-4354. [PMID: 32402755 PMCID: PMC7408485 DOI: 10.1016/j.vaccine.2020.03.063] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/26/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022]
Abstract
Short repeat-region peptides from PvCSP on a VLP protect against malaria. The AGDR tetramer from PvCSP VK210 can, on a VLP, also protect against malaria. Full-length PvCSP is much less protective as a vaccine than truncated PvCSP. Region I and II peptides confer no protection against malaria presented on a VLP.
Vivax malaria is a major cause of morbidity and mortality worldwide, with several million clinical cases per year and 2.5 billion at risk of infection. A vaccine is urgently needed but the most advanced malaria vaccine, VMP001, confers only very low levels of protection against vivax malaria challenge in humans. VMP001 is based on the circumsporozoite protein (CSP) of Plasmodium vivax. Here a virus-like particle, Qβ, is used as a platform to generate very high levels of antibody against peptides from PvCSP in mice, in order to answer questions important to further development of P. vivax CSP (PvCSP) vaccines. Minimal peptides from the VK210 and VK247 allelic variants of PvCSP are found to be highly protective as Qβ-peptide vaccines, using transgenic P. berghei parasites expressing the homologous PvCSP allelic variant. A target of neutralising antibodies within the nonamer unit repeat of VK210, AGDR, is found, as a Qβ-peptide vaccine, to provide partial protection against malaria challenge, and enhances protective efficacy when combined with full-length PvCSP vaccination. A truncated form of PvCSP, missing the N-terminal domain, is found to confer much higher levels of protective efficacy than full-length PvCSP. Peptides derived from highly conserved areas of PvCSP, RI and RII, are found not to confer protective efficacy as Qβ-peptide vaccines.
Collapse
|
5
|
Khan S, Parrillo M, Gutierrez AH, Terry FE, Moise L, Martin WD, De Groot AS. Immune escape and immune camouflage may reduce the efficacy of RTS,S vaccine in Malawi. Hum Vaccin Immunother 2019; 16:214-227. [PMID: 30614773 PMCID: PMC7062414 DOI: 10.1080/21645515.2018.1560772] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The RTS,S/AS01 malaria vaccine will undergo a pilot vaccination study in sub-Saharan Africa beginning in 2019. RTS,S/AS01 Phase III trials reported an efficacy of 28.3% (children 5–17 months) and 18.3% (infants 6–12 weeks), with substantial variability across study sites. We postulated that the relatively low efficacy of the RTS,S vaccine and variability across sites may be due to lack of T-cell epitopes in the vaccine antigen, and due to the HLA distribution of the vaccinated population, and/or due to ‘immune camouflage’, an immune escape mechanism. To examine these hypotheses, we used immunoinformatics tools to compare T helper epitopes contained in RTS,S vaccine antigens with Plasmodium falciparum circumsporozoite protein (CSP) variants isolated from infected individuals in Malawi. The prevalence of epitopes restricted by specific HLA-DRB1 alleles was inversely associated with prevalence of the HLA-DRB1 allele in the Malawi study population, suggesting immune escape. In addition, T-cell epitopes in the CSP of strains circulating in Malawi were more often restricted by low-frequency HLA-DRB1 alleles in the population. Furthermore, T-cell epitopes that were highly conserved across CSP variants in Malawi possessed TCR-facing residues that were highly conserved in the human proteome, potentially reducing T-cell help through tolerance. The CSP component of the RTS,S vaccine also exhibited a low degree of T-cell epitope relatedness to circulating variants. These results suggest that RTS,S vaccine efficacy may be impacted by low T-cell epitope content, reduced presentation of T-cell epitopes by prevalent HLA-DRB1, high potential for human-cross-reactivity, and limited conservation with the CSP of circulating malaria strains.
Collapse
Affiliation(s)
- Sundos Khan
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, USA
| | - Matthew Parrillo
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, USA
| | | | | | - Leonard Moise
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, USA.,EpiVax, Inc., Providence, RI, USA
| | | | - Anne S De Groot
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, USA.,EpiVax, Inc., Providence, RI, USA
| |
Collapse
|
6
|
Aliprandini E, Tavares J, Panatieri RH, Thiberge S, Yamamoto MM, Silvie O, Ishino T, Yuda M, Dartevelle S, Traincard F, Boscardin SB, Amino R. Cytotoxic anti-circumsporozoite antibodies target malaria sporozoites in the host skin. Nat Microbiol 2018; 3:1224-1233. [PMID: 30349082 DOI: 10.1038/s41564-018-0254-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 08/28/2018] [Indexed: 01/09/2023]
Abstract
The circumsporozoite protein (CSP) is the major surface protein of malaria sporozoites (SPZs), the motile and invasive parasite stage inoculated in the host skin by infected mosquitoes. Antibodies against the central CSP repeats of different plasmodial species are known to block SPZ infectivity1-5, but the precise mechanism by which these effectors operate is not completely understood. Here, using a rodent Plasmodium yoelii malaria model, we show that sterile protection mediated by anti-P. yoelii CSP humoral immunity depends on the parasite inoculation into the host skin, where antibodies inhibit motility and kill P. yoelii SPZs via a characteristic 'dotty death' phenotype. Passive transfer of an anti-repeat monoclonal antibody (mAb) recapitulates the skin inoculation-dependent protection, in a complement- and Fc receptor γ-independent manner. This purified mAb also decreases motility and, notably, induces the dotty death of P. yoelii SPZs in vitro. Cytotoxicity is species-transcendent since cognate anti-CSP repeat mAbs also kill Plasmodium berghei and Plasmodium falciparum SPZs. mAb cytotoxicity requires the actomyosin motor-dependent translocation and stripping of the protective CSP surface coat, rendering the parasite membrane susceptible to the SPZ pore-forming-like protein secreted to wound and traverse the host cell membrane6. The loss of SPZ fitness caused by anti-P. yoelii CSP repeat antibodies is thus a dynamic process initiated in the host skin where SPZs either stop moving7, or migrate and traverse cells to progress through the host tissues7-9 at the eventual expense of their own life.
Collapse
Affiliation(s)
| | - Joana Tavares
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Raquel Hoffmann Panatieri
- Unit of Malaria Infection & Immunity, Institut Pasteur, Paris, France.,Parasitology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sabine Thiberge
- Unit of Malaria Infection & Immunity, Institut Pasteur, Paris, France.,Centre de Production et d'Infection des Anophèles, Institut Pasteur, Paris, France
| | - Marcio Massao Yamamoto
- Parasitology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Olivier Silvie
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI, Paris, France
| | - Tomoko Ishino
- Department of Molecular Parasitology, Ehime University, Ehime, Japan
| | - Masao Yuda
- Department of Medical Zoology, Mie University School of Medicine, Mie, Japan
| | - Sylvie Dartevelle
- Plateforme d'Ingénierie des Anticorps, Institut Pasteur, Paris, France
| | | | - Silvia Beatriz Boscardin
- Parasitology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | - Rogerio Amino
- Unit of Malaria Infection & Immunity, Institut Pasteur, Paris, France.
| |
Collapse
|
7
|
A public antibody lineage that potently inhibits malaria infection through dual binding to the circumsporozoite protein. Nat Med 2018; 24:401-407. [PMID: 29554084 PMCID: PMC5893353 DOI: 10.1038/nm.4513] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 02/08/2018] [Indexed: 12/13/2022]
Abstract
Immunization with attenuated Plasmodium falciparum sporozoites (PfSPZs) has been shown to be protective against malaria, but the features of the antibody response induced by this treatment remain unclear. To investigate this response in detail, we isolated IgM and IgG monoclonal antibodies from Tanzanian volunteers who were immunized with repeated injection of Sanaria PfSPZ Vaccine and who were found to be protected from controlled human malaria infection with infectious homologous PfSPZs. All isolated IgG monoclonal antibodies bound to P. falciparum circumsporozoite protein (PfCSP) and recognized distinct epitopes in its N terminus, NANP-repeat region, and C terminus. Strikingly, the most effective antibodies, as determined in a humanized mouse model, bound not only to the repeat region, but also to a minimal peptide at the PfCSP N-terminal junction that is not in the RTS,S vaccine. These dual-specific antibodies were isolated from different donors and were encoded by VH3-30 or VH3-33 alleles that encode tryptophan or arginine at position 52. Using structural and mutational data, we describe the elements required for germline recognition and affinity maturation. Our study provides potent neutralizing antibodies and relevant information for lineage-targeted vaccine design and immunization strategies.
Collapse
|
8
|
Murugan R, Buchauer L, Triller G, Kreschel C, Costa G, Pidelaserra Martí G, Imkeller K, Busse CE, Chakravarty S, Sim BKL, Hoffman SL, Levashina EA, Kremsner PG, Mordmüller B, Höfer T, Wardemann H. Clonal selection drives protective memory B cell responses in controlled human malaria infection. Sci Immunol 2018; 3:3/20/eaap8029. [DOI: 10.1126/sciimmunol.aap8029] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/30/2017] [Indexed: 01/20/2023]
|
9
|
Scally SW, Murugan R, Bosch A, Triller G, Costa G, Mordmüller B, Kremsner PG, Sim BKL, Hoffman SL, Levashina EA, Wardemann H, Julien JP. Rare PfCSP C-terminal antibodies induced by live sporozoite vaccination are ineffective against malaria infection. J Exp Med 2017; 215:63-75. [PMID: 29167197 PMCID: PMC5748854 DOI: 10.1084/jem.20170869] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 08/30/2017] [Accepted: 10/11/2017] [Indexed: 11/16/2022] Open
Abstract
Scally et al. show the molecular, structural, and functional characterization of human antibodies against the C-terminal domain of Plasmodium falciparum (Pf) circumsporozoite (CSP [C-PfCSP]) and reveal that its arrangement on the Pf sporozoite surface and epitope polymorphism contribute to poor C-PfCSP immunogenicity and ineffective humoral responses in volunteers protected against Pf malaria. Antibodies against the central repeat of the Plasmodium falciparum (Pf) circumsporozoite protein (CSP) inhibit parasite activity and correlate with protection from malaria. However, the humoral response to the PfCSP C terminus (C-PfCSP) is less well characterized. Here, we describe B cell responses to C-PfCSP from European donors who underwent immunization with live Pf sporozoites (PfSPZ Challenge) under chloroquine prophylaxis (PfSPZ-CVac), and were protected against controlled human malaria infection. Out of 215 PfCSP-reactive monoclonal antibodies, only two unique antibodies were specific for C-PfCSP, highlighting the rare occurrence of C-PfCSP–reactive B cells in PfSPZ-CVac–induced protective immunity. These two antibodies showed poor sporozoite binding and weak inhibition of parasite traversal and development, and did not protect mice from infection with PfCSP transgenic Plasmodium berghei sporozoites. Structural analyses demonstrated that one antibody interacts with a polymorphic region overlapping two T cell epitopes, suggesting that variability in C-PfCSP may benefit parasite escape from humoral and cellular immunity. Our data identify important features underlying C-PfCSP shortcomings as a vaccine target.
Collapse
Affiliation(s)
- Stephen W Scally
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Rajagopal Murugan
- B Cell Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Alexandre Bosch
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Gianna Triller
- B Cell Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Giulia Costa
- Vector Biology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Benjamin Mordmüller
- Institute of Tropical Medicine and German Center for Infection Research, University of Tübingen, Tübingen, Germany
| | - Peter G Kremsner
- Institute of Tropical Medicine and German Center for Infection Research, University of Tübingen, Tübingen, Germany
| | | | | | - Elena A Levashina
- Vector Biology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Hedda Wardemann
- B Cell Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Jean-Philippe Julien
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada .,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Thomas BE, Sridevi K, Chopra N, Haq W, Rao DN. Inducing a cell-mediated immune response against peptides of thePlasmodium vivaxcircumsporozoite protein. ANNALS OF TROPICAL MEDICINE AND PARASITOLOGY 2016. [DOI: 10.1080/00034983.2001.11813672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
11
|
Arévalo-Herrera M, Roggero MA, Gonzalez JM, Vergara J, Corradin G, López JA, Herrera S. Mapping and comparison of the B-cell epitopes recognized on thePlasmodium vivaxcircumsporozoite protein by immune Colombians and immunizedAotusmonkeys. ANNALS OF TROPICAL MEDICINE AND PARASITOLOGY 2016. [DOI: 10.1080/00034983.1998.11813311] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
12
|
Thomas BE, Manocha M, Haq W, Adak T, Pillai CR, Rao DN. Modulation of the humoral response to repeat and non-repeat sequences of the circumsporozoite protein ofPlasmodium vivaxusing novel adjuvant and delivery systems. ANNALS OF TROPICAL MEDICINE AND PARASITOLOGY 2016. [DOI: 10.1080/00034983.2001.11813659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
13
|
Ishizuka AS, Lyke KE, DeZure A, Berry AA, Richie TL, Mendoza FH, Enama ME, Gordon IJ, Chang LJ, Sarwar UN, Zephir KL, Holman LA, James ER, Billingsley PF, Gunasekera A, Chakravarty S, Manoj A, Li M, Ruben AJ, Li T, Eappen AG, Stafford RE, K C N, Murshedkar T, DeCederfelt H, Plummer SH, Hendel CS, Novik L, Costner PJM, Saunders JG, Laurens MB, Plowe CV, Flynn B, Whalen WR, Todd JP, Noor J, Rao S, Sierra-Davidson K, Lynn GM, Epstein JE, Kemp MA, Fahle GA, Mikolajczak SA, Fishbaugher M, Sack BK, Kappe SHI, Davidson SA, Garver LS, Björkström NK, Nason MC, Graham BS, Roederer M, Sim BKL, Hoffman SL, Ledgerwood JE, Seder RA. Protection against malaria at 1 year and immune correlates following PfSPZ vaccination. Nat Med 2016; 22:614-23. [PMID: 27158907 PMCID: PMC11294733 DOI: 10.1038/nm.4110] [Citation(s) in RCA: 273] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 04/15/2016] [Indexed: 02/07/2023]
Abstract
An attenuated Plasmodium falciparum (Pf) sporozoite (SPZ) vaccine, PfSPZ Vaccine, is highly protective against controlled human malaria infection (CHMI) 3 weeks after immunization, but the durability of protection is unknown. We assessed how vaccine dosage, regimen, and route of administration affected durable protection in malaria-naive adults. After four intravenous immunizations with 2.7 × 10(5) PfSPZ, 6/11 (55%) vaccinated subjects remained without parasitemia following CHMI 21 weeks after immunization. Five non-parasitemic subjects from this dosage group underwent repeat CHMI at 59 weeks, and none developed parasitemia. Although Pf-specific serum antibody levels correlated with protection up to 21-25 weeks after immunization, antibody levels waned substantially by 59 weeks. Pf-specific T cell responses also declined in blood by 59 weeks. To determine whether T cell responses in blood reflected responses in liver, we vaccinated nonhuman primates with PfSPZ Vaccine. Pf-specific interferon-γ-producing CD8 T cells were present at ∼100-fold higher frequencies in liver than in blood. Our findings suggest that PfSPZ Vaccine conferred durable protection to malaria through long-lived tissue-resident T cells and that administration of higher doses may further enhance protection.
Collapse
Affiliation(s)
- Andrew S Ishizuka
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Kirsten E Lyke
- Institute for Global Health, Center for Vaccine Development and Division of Malaria Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Adam DeZure
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Andrea A Berry
- Institute for Global Health, Center for Vaccine Development and Division of Malaria Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Floreliz H Mendoza
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Mary E Enama
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Ingelise J Gordon
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Lee-Jah Chang
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Uzma N Sarwar
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Kathryn L Zephir
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - LaSonji A Holman
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | | | | | | | | | | | - MingLin Li
- Sanaria Inc., Rockville, Maryland, USA
- Protein Potential, LLC, Rockville, Maryland, USA
| | | | - Tao Li
- Sanaria Inc., Rockville, Maryland, USA
| | | | - Richard E Stafford
- Sanaria Inc., Rockville, Maryland, USA
- Protein Potential, LLC, Rockville, Maryland, USA
| | - Natasha K C
- Sanaria Inc., Rockville, Maryland, USA
- Protein Potential, LLC, Rockville, Maryland, USA
| | | | - Hope DeCederfelt
- Pharmaceutical Development Section, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Sarah H Plummer
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Cynthia S Hendel
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Laura Novik
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Pamela J M Costner
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Jamie G Saunders
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Matthew B Laurens
- Institute for Global Health, Center for Vaccine Development and Division of Malaria Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Christopher V Plowe
- Institute for Global Health, Center for Vaccine Development and Division of Malaria Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Barbara Flynn
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - William R Whalen
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - J P Todd
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Jay Noor
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Srinivas Rao
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Kailan Sierra-Davidson
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Geoffrey M Lynn
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Judith E Epstein
- Naval Medical Research Center (NMRC), Malaria Department, Silver Spring, Maryland, USA
| | - Margaret A Kemp
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Gary A Fahle
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - Brandon K Sack
- Center for Infectious Disease Research, Seattle, Washington, USA
| | - Stefan H I Kappe
- Center for Infectious Disease Research, Seattle, Washington, USA
| | - Silas A Davidson
- Entomology Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Lindsey S Garver
- Entomology Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Niklas K Björkström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Martha C Nason
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Barney S Graham
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Mario Roederer
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - B Kim Lee Sim
- Sanaria Inc., Rockville, Maryland, USA
- Protein Potential, LLC, Rockville, Maryland, USA
| | | | - Julie E Ledgerwood
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Robert A Seder
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| |
Collapse
|
14
|
Yadava A, Hall CE, Sullivan JS, Nace D, Williams T, Collins WE, Ockenhouse CF, Barnwell JW. Protective efficacy of a Plasmodium vivax circumsporozoite protein-based vaccine in Aotus nancymaae is associated with antibodies to the repeat region. PLoS Negl Trop Dis 2014; 8:e3268. [PMID: 25329054 PMCID: PMC4199514 DOI: 10.1371/journal.pntd.0003268] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 09/12/2014] [Indexed: 11/18/2022] Open
Abstract
We have previously reported that Vivax Malaria Protein 001 (VMP001), a vaccine candidate based on the circumsporozoite protein of Plasmodium vivax, is immunogenic in mice and rhesus monkeys in the presence of various adjuvants. In the present study, we evaluated the immunogenicity and efficacy of VMP001 formulated with a TLR9 agonist in a water-in-oil emulsion. Following immunization, the vaccine efficacy was assessed by challenging Aotus nancymaae monkeys with P. vivax sporozoites. Monkeys from both the low- and high-dose vaccine groups generated strong humoral immune responses to the vaccine (peak median titers of 291,622), and its subunits (peak median titers to the N-term, central repeat and C-term regions of 22,188; 66,120 and 179,947, respectively). 66.7% of vaccinated monkeys demonstrated sterile protection following challenge. Protection was associated with antibodies directed against the central repeat region. The protected monkeys had a median anti-repeat titer of 97,841 compared to 14,822 in the non-protected monkeys. This is the first report demonstrating P. vivax CSP vaccine-induced protection of Aotus monkeys challenged with P. vivax sporozoites. Plasmodium vivax is responsible for causing malaria in large parts of the globe, including regions with temperate climates not suited for the transmission of other Plasmodium species. In addition, P. vivax has the propensity to form dormant forms, known as hypnozoites, that can remain latent for weeks to months and reactive periodically to cause recurrent infections. Prevention of P. vivax malaria, more than any other form, will require a vaccine-based intervention due to limitations in treatment options. To this end, we tested the efficacy in non-human primates, of a vaccine based on circumsporozoite protein, a preerythrocytic stage antigen, of P. vivax. Aotus monkeys were immunized with clinical-grade antigen, combined with two immunomodulators, and then challenged with P. vivax sporozoites. Following challenge 66.7% of monkeys were protected. Analysis of serum samples indicated that protection was associated with antibodies to the central repeat region of the molecule, and that protection was lost upon waning of these antibodies. This is the first report demonstrating that active immunization with a recombinant protein can lead to complete protection in monkeys following sporozoite challenge, while also demonstrating a protective associate. Our data can help serve as a benchmark for down-selection of future vaccine formulations for P. vivax.
Collapse
Affiliation(s)
- Anjali Yadava
- Malaria Vaccine Branch, Military Malaria Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- * E-mail:
| | - Cysha E. Hall
- Malaria Vaccine Branch, Military Malaria Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - JoAnn S. Sullivan
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Douglas Nace
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Tyrone Williams
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - William E. Collins
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Christian F. Ockenhouse
- Malaria Vaccine Branch, Military Malaria Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - John W. Barnwell
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| |
Collapse
|
15
|
Vanloubbeeck Y, Pichyangkul S, Bayat B, Yongvanitchit K, Bennett JW, Sattabongkot J, Schaecher K, Ockenhouse CF, Cohen J, Yadava A. Comparison of the immune responses induced by soluble and particulate Plasmodium vivax circumsporozoite vaccine candidates formulated in AS01 in rhesus macaques. Vaccine 2013; 31:6216-24. [DOI: 10.1016/j.vaccine.2013.10.041] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/26/2013] [Accepted: 10/08/2013] [Indexed: 11/26/2022]
|
16
|
Cheng Y, Ito D, Sattabongkot J, Lim CS, Kong DH, Ha KS, Wang B, Tsuboi T, Han ET. Serological responses to a soluble recombinant chimeric Plasmodium vivax circumsporozoite protein in VK210 and VK247 population. Malar J 2013; 12:323. [PMID: 24034268 PMCID: PMC3847697 DOI: 10.1186/1475-2875-12-323] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 09/11/2013] [Indexed: 12/01/2022] Open
Abstract
Background Circumsporozoite protein (CSP) is essential for sporozoite formation and sporozoite invasion into human hepatocyte. Previously, a recombinant P. vivax CSP based on chimeric repeats (rPvCSP-c) representing two major alleles VK210 and VK247 within central region has been designed. Naturally acquired humoral immune responses study show that antigenicity of rPvCSP-c was much higher than that of native strain. However, the serologic reactivity of rPvCSP-c was still unclear in detail. Methods In present study, recognition of rPvCSP-c in vivax malaria typed VK210 and VK247 alleles was assessed. VK210 typed and VK247 typed sera from adult residents reacted specifically with rPvCSP-c using protein array and immunoblot assay. Additionally, anti-rPvCSP-c serum recognized the fixed VK210 and VK247 sporozoites by immunofluorescence assay. Furthermore, statistic analysis was performed for correlational detection. Results The rPvCSP-c reacted with both VK210 typed and VK247 typed P. vivax infected patient sera and anti-rPvCSP-c immune serum also reacted with VK210 and VK247 sporozoite parasites of P. vivax specifically. There was a positive correlation between increased antibody level, age of patients and also associated with pvcsp repeat number, although the level of responses did vary considerably in their reactivity to the rPvCSP-c from negative to very high level within each age group. Conclusions These data confirmed the serologic reactivity of the novel rPvCSP-c in exposed both VK210 and VK247 populations. These results strongly suggested that this recombinant CSP was biologically active and potently immunogenic across major strains and raised the prospect that this protein could be used as serologic marker.
Collapse
Affiliation(s)
- Yang Cheng
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 200-701, Republic of Korea.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Immunogenicity of recombinant proteins consisting of Plasmodium vivax circumsporozoite protein allelic variant-derived epitopes fused with Salmonella enterica Serovar Typhimurium flagellin. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 20:1418-25. [PMID: 23863502 DOI: 10.1128/cvi.00312-13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A Plasmodium falciparum circumsporozoite protein (CSP)-based recombinant fusion vaccine is the first malaria vaccine to reach phase III clinical trials. Resistance to infection correlated with the production of antibodies to the immunodominant central repeat region of the CSP. In contrast to P. falciparum, vaccine development against the CSP of Plasmodium vivax malaria is far behind. Based on this gap in our knowledge, we generated a recombinant chimeric protein containing the immunodominant central repeat regions of the P. vivax CSP fused to Salmonella enterica serovar Typhimurium-derived flagellin (FliC) to activate the innate immune system. The recombinant proteins that were generated contained repeat regions derived from each of the 3 different allelic variants of the P. vivax CSP or a fusion of regions derived from each of the 3 allelic forms. Mice were subcutaneously immunized with the fusion proteins alone or in combination with the Toll-like receptor 3 (TLR-3) agonist poly(I·C), and the anti-CSP serum IgG response was measured. Immunization with a mixture of the 3 recombinant proteins, each containing immunodominant epitopes derived from a single allelic variant, rather than a single recombinant protein carrying a fusion of regions derived from each of 3 allelic forms elicited a stronger immune response. This response was independent of TLR-4 but required TLR-5/MyD88 activation. Antibody titers significantly increased when poly(I·C) was used as an adjuvant with a mixture of the 3 recombinant proteins. These recombinant fusion proteins are novel candidates for the development of an effective malaria vaccine against P. vivax.
Collapse
|
18
|
Moon JJ, Suh H, Polhemus ME, Ockenhouse CF, Yadava A, Irvine DJ. Antigen-displaying lipid-enveloped PLGA nanoparticles as delivery agents for a Plasmodium vivax malaria vaccine. PLoS One 2012; 7:e31472. [PMID: 22328935 PMCID: PMC3273465 DOI: 10.1371/journal.pone.0031472] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 01/11/2012] [Indexed: 11/18/2022] Open
Abstract
The parasite Plasmodium vivax is the most frequent cause of malaria outside of sub-Saharan Africa, but efforts to develop viable vaccines against P. vivax so far have been inadequate. We recently developed pathogen-mimicking polymeric vaccine nanoparticles composed of the FDA-approved biodegradable polymer poly(lactide-co-glycolide) acid (PLGA) “enveloped” by a lipid membrane. In this study, we sought to determine whether this vaccine delivery platform could be applied to enhance the immune response against P. vivax sporozoites. A candidate malaria antigen, VMP001, was conjugated to the lipid membrane of the particles, and an immunostimulatory molecule, monophosphoryl lipid A (MPLA), was incorporated into the lipid membranes, creating pathogen-mimicking nanoparticle vaccines (VMP001-NPs). Vaccination with VMP001-NPs promoted germinal center formation and elicited durable antigen-specific antibodies with significantly higher titers and more balanced Th1/Th2 responses in vivo, compared with vaccines composed of soluble protein mixed with MPLA. Antibodies raised by NP vaccinations also exhibited enhanced avidity and affinity toward the domains within the circumsporozoite protein implicated in protection and were able to agglutinate live P. vivax sporozoites. These results demonstrate that these VMP001-NPs are promising vaccines candidates that may elicit protective immunity against P. vivax sporozoites.
Collapse
Affiliation(s)
- James J. Moon
- Department of Materials Science and Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, United States of America
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, United States of America
| | - Heikyung Suh
- Department of Materials Science and Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, United States of America
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Mark E. Polhemus
- Division of Malaria Vaccine Development, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Christian F. Ockenhouse
- Division of Malaria Vaccine Development, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Anjali Yadava
- Division of Malaria Vaccine Development, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Darrell J. Irvine
- Department of Materials Science and Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, United States of America
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, United States of America
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, United States of America
- Ragon Institute of MGH, MIT, and Harvard, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
- * E-mail:
| |
Collapse
|
19
|
Enhancing humoral responses to a malaria antigen with nanoparticle vaccines that expand Tfh cells and promote germinal center induction. Proc Natl Acad Sci U S A 2012; 109:1080-5. [PMID: 22247289 DOI: 10.1073/pnas.1112648109] [Citation(s) in RCA: 263] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
For subunit vaccines, adjuvants play a key role in shaping immunological memory. Nanoparticle (NP) delivery systems for antigens and/or molecular danger signals are promising adjuvants capable of promoting both cellular and humoral immune responses, but in most cases the mechanisms of action of these materials are poorly understood. Here, we studied the immune response elicited by NPs composed of multilamellar "stapled" lipid vesicles carrying a recombinant Plasmodium vivax circumsporozoite antigen, VMP001, both entrapped in the aqueous core and anchored to the lipid bilayer surfaces. Immunization with these particles and monophosphoryl lipid A (MPLA), a US Food and Drug Administration-approved immunostimulatory agonist for Toll-like receptor-4, promoted high-titer, high-avidity antibody responses against VMP001, lasting more than 1 y in mice at 10-fold lower doses than conventional adjuvants. Compared to soluble VMP001 mixed with MPLA, VMP001-NPs promoted broader humoral responses, targeting multiple epitopes of the protein and a more balanced Th1/Th2 cytokine profile from antigen-specific T cells. To begin to understand the underlying mechanisms, we examined components of the B-cell response and found that NPs promoted robust germinal center (GC) formation at low doses of antigen where no GC induction occurred with soluble protein immunization, and that GCs nucleated near depots of NPs accumulating in the draining lymph nodes over time. In parallel, NP vaccination enhanced the expansion of antigen-specific follicular helper T cells (T(fh)), compared to vaccinations with soluble VMP001 or alum. Thus, NP vaccines may be a promising strategy to enhance the durability, breadth, and potency of humoral immunity by enhancing key elements of the B-cell response.
Collapse
|
20
|
Evaluation of the safety and immunogenicity in rhesus monkeys of a recombinant malaria vaccine for Plasmodium vivax with a synthetic Toll-like receptor 4 agonist formulated in an emulsion. Infect Immun 2011; 79:3492-500. [PMID: 21690242 DOI: 10.1128/iai.05257-11] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Plasmodium vivax is the major cause of malaria outside sub-Saharan Africa and inflicts debilitating morbidity and consequent economic impacts in developing countries. In order to produce a P. vivax vaccine for global use, we have previously reported the development of a novel chimeric recombinant protein, VMP001, based on the circumsporozoite protein (CSP) of P. vivax. Very few adjuvant formulations are currently available for human use. Our interest is to evaluate second-generation vaccine formulations to identify novel combinations of adjuvants capable of inducing strong, long-lasting immune responses. In this study rhesus monkeys were immunized intramuscularly three times with VMP001 in combination with a stable emulsion (SE) or a synthetic Toll-like receptor 4 (TLR4) agonist (glucopyranosyl lipid A [GLA]) in SE (GLA-SE). Sera and peripheral blood mononuclear cells (PBMCs) were tested for the presence of antigen-specific humoral and cellular responses, respectively. All groups of monkeys generated high titers of anti-P. vivax IgG antibodies, as detected by enzyme-linked immunosorbent assays (ELISAs) and immunofluorescence assays. In addition, all groups generated a cellular immune response characterized by antigen-specific CD4(+) T cells secreting predominantly interleukin-2 (IL-2) and lesser amounts of tumor necrosis factor (TNF). We conclude that the combination of VMP001 and GLA-SE is safe and immunogenic in monkeys and may serve as a potential second-generation vaccine candidate against P. vivax malaria.
Collapse
|
21
|
Establishment of an in vitro assay for assessing the effects of drugs on the liver stages of Plasmodium vivax malaria. PLoS One 2010; 5:e14275. [PMID: 21151554 PMCID: PMC3000326 DOI: 10.1371/journal.pone.0014275] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 11/17/2010] [Indexed: 11/19/2022] Open
Abstract
Plasmodium vivax (Pv) is the second most important human malaria parasite. Recent data indicate that the impact of Pv malaria on the health and economies of the developing world has been dramatically underestimated. Pv has a unique feature in its life cycle. Uninucleate sporozoites (spz), after invasion of human hepatocytes, either proceed to develop into tens of thousands of merozoites within the infected hepatocytes or remain as dormant forms called hypnozoites, which cause relapses of malaria months to several years after the primary infection. Elimination of malaria caused by Pv will be facilitated by developing a safe, highly effective drug that eliminates Pv liver stages, including hypnozoites. Identification and development of such a drug would be facilitated by the development of a medium to high throughput assay for screening drugs against Pv liver stages. We undertook the present pilot study to (1) assess the feasibility of producing large quantities of purified, vialed, cryopreserved Pv sporozoites and (2) establish a system for culturing the liver stages of Pv in order to assess the effects of drugs on the liver stages of Pv. We used primaquine (PQ) to establish this assay model, because PQ is the only licensed drug known to clear all Pv hepatocyte stages, including hypnozoites, and the effect of PQ on Pv hepatocyte stage development in vitro has not previously been reported. We report that we have established the capacity to reproducibly infect hepatoma cells with purified, cyropreserved Pv spz from the same lot, quantitate the primary outcome variable of infected hepatoma cells and demonstrate the inhibitory activity of primaquine on the infected hepatoma cells. We have also identified small parasite forms that may be hypnozoites. These data provide the foundation for finalizing a medium throughput, high content assay to identify new drugs for the elimination of all Pv liver stages.
Collapse
|
22
|
Guilbride DL, Gawlinski P, Guilbride PDL. Why functional pre-erythrocytic and bloodstage malaria vaccines fail: a meta-analysis of fully protective immunizations and novel immunological model. PLoS One 2010; 5:e10685. [PMID: 20502667 PMCID: PMC2873430 DOI: 10.1371/journal.pone.0010685] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 04/16/2010] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Clinically protective malaria vaccines consistently fail to protect adults and children in endemic settings, and at best only partially protect infants. METHODOLOGY/PRINCIPAL FINDINGS We identify and evaluate 1916 immunization studies between 1965-February 2010, and exclude partially or nonprotective results to find 177 completely protective immunization experiments. Detailed reexamination reveals an unexpectedly mundane basis for selective vaccine failure: live malaria parasites in the skin inhibit vaccine function. We next show published molecular and cellular data support a testable, novel model where parasite-host interactions in the skin induce malaria-specific regulatory T cells, and subvert early antigen-specific immunity to parasite-specific immunotolerance. This ensures infection and tolerance to reinfection. Exposure to Plasmodium-infected mosquito bites therefore systematically triggers immunosuppression of endemic vaccine-elicited responses. The extensive vaccine trial data solidly substantiate this model experimentally. CONCLUSIONS/SIGNIFICANCE We conclude skinstage-initiated immunosuppression, unassociated with bloodstage parasites, systematically blocks vaccine function in the field. Our model exposes novel molecular and procedural strategies to significantly and quickly increase protective efficacy in both pipeline and currently ineffective malaria vaccines, and forces fundamental reassessment of central precepts determining vaccine development. This has major implications for accelerated local eliminations of malaria, and significantly increases potential for eradication.
Collapse
|
23
|
Good MF. The hope but challenge for developing a vaccine that might control malaria. Eur J Immunol 2009; 39:939-43. [DOI: 10.1002/eji.200939390] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
24
|
Abstract
More attention is being focused on malaria today than any time since the world's last efforts to achieve eradication over 40 years ago. The global community is now discussing strategies aimed at dramatically reducing malarial disease burden and the eventual eradication of all types of malaria, everywhere. As a consequence, Plasmodium vivax, which has long been neglected and mistakenly considered inconsequential, is now entering into the strategic debates taking place on malaria epidemiology and control, drug resistance, pathogenesis and vaccines. Thus, contrary to the past, the malaria research community is becoming more aware and concerned about the widespread spectrum of illness and death caused by up to a couple of hundred million cases of vivax malaria each year. This review brings these issues to light and provides an overview of P. vivax vaccine development, then and now. Progress had been slow, given inherent research challenges and minimal support in the past, but prospects are looking better for making headway in the next few years. P. vivax, known to invade the youngest red blood cells, the reticulocytes, presents a strong challenge towards developing a reliable long-term culture system to facilitate needed research. The P. vivax genome was published recently, and vivax researchers now need to coordinate efforts to discover new vaccine candidates, establish new vaccine approaches, capitalize on non-human primate models for testing, and investigate the unique biological features of P. vivax, including the elusive P. vivax hypnozoites. Comparative studies on both P. falciparum and P. vivax in many areas of research will be essential to eradicate malaria. And to this end, the education and training of future generations of dedicated "malariologists" to advance our knowledge, understanding and the development of new interventions against each of the malaria species infecting humans also will be essential.
Collapse
Affiliation(s)
- Mary R Galinski
- Emory Vaccine Center and Yerkes National Primate Research Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - John W Barnwell
- Malaria Branch, Division of Parasitic Diseases, National Center for Zoonotic, Vector-Borne and Enteric Diseases, the Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
25
|
Abstract
Plasmodium vivax is a significant public health threat throughout most of the tropics and to travelers to these regions. The infection causes a debilitating febrile syndrome that often recurs and in rare cases ends in death. The complex life cycle of the parasite compounds the difficulty of prevention and treatment, principally due to the phenomenon of relapse. Most commonly used drugs for preventing malaria fail to prevent late relapses by this parasite. Treatment requires dealing with both blood and liver stages. Since 1950, primaquine has been the only drug available for treatment of liver stages, and important clinical questions surround its appropriate use (ie, dosing, efficacy, safety, and tolerability). Likewise, chloroquine has been first-line therapy for vivax malaria since 1946, and the emergence of resistance to the drug further complicates therapeutic management decisions.
Collapse
Affiliation(s)
- J Kevin Baird
- ALERTAsia Foundation, c/o Eijkman Institute for Molecular Biology, Jalan Diponegoro No. 69, Menteng, Jakarta 10430, Indonesia.
| | | | | |
Collapse
|
26
|
Yadava A, Sattabongkot J, Washington MA, Ware LA, Majam V, Zheng H, Kumar S, Ockenhouse CF. A novel chimeric Plasmodium vivax circumsporozoite protein induces biologically functional antibodies that recognize both VK210 and VK247 sporozoites. Infect Immun 2006; 75:1177-85. [PMID: 17158893 PMCID: PMC1828583 DOI: 10.1128/iai.01667-06] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A successful vaccine against Plasmodium vivax malaria would significantly improve the health and quality of the lives of more than 1 billion people around the world. A subunit vaccine is the only option in the absence of long-term culture of P. vivax parasites. The circumsporozoite protein that covers the surface of Plasmodium sporozoites is one of the best-studied malarial antigens and the most promising vaccine in clinical trials. We report here the development of a novel "immunologically optimal" recombinant vaccine expressed in Escherichia coli that encodes a chimeric CS protein encompassing repeats from the two major alleles, VK210 and VK247. This molecule is widely recognized by sera from patients naturally exposed to P. vivax infection and induces a highly potent immune response in genetically disparate strains of mice. Antibodies from immunized animals recognize both VK210 and VK247 sporozoites. Furthermore, these antibodies appear to be protective in nature since they cause the agglutination of live sporozoites, an in vitro surrogate of sporozoite infectivity. These results strongly suggest that recombinant CS is biologically active and highly immunogenic across major histocompatibility complex strains and raises the prospect that in humans this vaccine may induce protective immune responses.
Collapse
Affiliation(s)
- Anjali Yadava
- Division of Malaria Vaccine Development, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Boscardin SB, Hafalla JCR, Masilamani RF, Kamphorst AO, Zebroski HA, Rai U, Morrot A, Zavala F, Steinman RM, Nussenzweig RS, Nussenzweig MC. Antigen targeting to dendritic cells elicits long-lived T cell help for antibody responses. ACTA ACUST UNITED AC 2006; 203:599-606. [PMID: 16505139 PMCID: PMC2118236 DOI: 10.1084/jem.20051639] [Citation(s) in RCA: 208] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Resistance to several prevalent infectious diseases requires both cellular and humoral immune responses. T cell immunity is initiated by mature dendritic cells (DCs) in lymphoid organs, whereas humoral responses to most antigens require further collaboration between primed, antigen-specific helper T cells and naive or memory B cells. To determine whether antigens delivered to DCs in lymphoid organs induce T cell help for antibody responses, we targeted a carrier protein, ovalbumin (OVA), to DCs in the presence of a maturation stimulus and assayed for antibodies to a hapten, (4-hydroxy-3-nitrophenyl) acetyl (NP), after boosting with OVA-NP. A single DC-targeted immunization elicited long-lived T cell helper responses to the carrier protein, leading to large numbers of antibody-secreting cells and high titers of high-affinity antihapten immunoglobulin Gs. Small doses of DC-targeted OVA induced higher titers and a broader spectrum of anti-NP antibody isotypes than large doses of OVA in alum adjuvant. Similar results were obtained when the circumsporozoite protein of Plasmodium yoelii was delivered to DCs. We conclude that antigen targeting to DCs combined with a maturation stimulus produces broad-based and long-lived T cell help for humoral immune responses.
Collapse
Affiliation(s)
- Silvia B Boscardin
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Herrera S, Bonelo A, Perlaza BL, Fernández OL, Victoria L, Lenis AM, Soto L, Hurtado H, Acuña LM, Vélez JD, Palacios R, Chen-Mok M, Corradin G, Arévalo-Herrera M. Safety and elicitation of humoral and cellular responses in colombian malaria-naive volunteers by a Plasmodium vivax circumsporozoite protein-derived synthetic vaccine. Am J Trop Med Hyg 2006; 73:3-9. [PMID: 16291760 DOI: 10.4269/ajtmh.2005.73.3] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Substantial experimental evidence indicates that the Plasmodium circumsporozoite (CS) protein has great potential as a vaccine candidate. We tested the safety and immunogenicity of vaccines composed of P. vivax CS-derived synthetic peptides. Sixty-nine healthy, malaria-naive volunteers were randomized to receive three injections of placebo or synthetic proteins N, R, or C (10, 30, or 100 microg/dose) in a double-blinded fashion. Vaccines were well tolerated and no serious adverse events were observed. Peptides N and R elicited humoral responses at all doses; peptide C elicicted these responses only at doses of 30 and 100 microg. The N peptide at a dose of 100 microg elicited the greatest antibody response. Antibodies to the three peptides recognized P. vivax sporozoites in an immunofluorescent antibody test. Peripheral blood mononuclear cells from most immunized volunteers also produced interferon-gamma upon peptide in vitro stimulation. These vaccines appear safe, well tolerated, and immunogenic in malaria-naive volunteers. Further optimization and development of this vaccine is being attempted to conduct phase II clinical trials.
Collapse
Affiliation(s)
- Sócrates Herrera
- Instituto de Inmunología, Universidad del Valle, Cali, Colombia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Herrera S, Bonelo A, Perlaza BL, Valencia AZ, Cifuentes C, Hurtado S, Quintero G, López JA, Corradin G, Arévalo-Herrera M. Use of long synthetic peptides to study the antigenicity and immunogenicity of the Plasmodium vivax circumsporozoite protein. Int J Parasitol 2005; 34:1535-46. [PMID: 15582530 DOI: 10.1016/j.ijpara.2004.10.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2004] [Revised: 10/04/2004] [Accepted: 10/05/2004] [Indexed: 11/19/2022]
Abstract
Three long synthetic peptides corresponding to amino (N), repeat (R) and carboxyl (C) regions of the Plasmodium vivax circumsporozoite (CS) protein were synthesised and used to assess their potential as vaccine candidates. Antigenicity studies were carried out using human blood samples from residents of a malaria-endemic area of Colombia, and immunogenicity was tested in Aotus monkeys. The N and C peptides spanned the total native amino and carboxyl flanking regions, whereas the R peptide corresponded to a construct based on the first central nona-peptide repeated in tandem three times and colinearly linked to a universal T-cell epitope (ptt-30) derived from tetanus toxin. All three peptides had been shown previously to contain several B-, T-helper (Th) and Cytotoxic T Lymphocytes (CTL) epitopes. Sixty-one percent of the human sera reacted with the R region, whereas 35 and 39% of the samples had antibodies against the N and C peptides, respectively. Human Peripheral Blood Mononuclear Cells (PBMC) showed higher levels of IFN-gamma than IL-4 when stimulated with peptides containing Th epitopes. Aotus monkeys immunised with the peptides formulated in either Montanide ISA720 or Freund's adjuvants produced strong antibody responses that recognised the peptide immunogens and the native circumsporozoite protein on sporozoites. Additionally, high IFN-gamma production was induced when Aotus lymphocytes were stimulated in vitro with each of the three peptides. We observed boosting of antibody responses and IFN-gamma production by exposure to live sporozoites. These results confirm the high antigenicity and immunogenicity of such synthetic polypeptides and underline their vaccine potential.
Collapse
Affiliation(s)
- Sócrates Herrera
- Institute of Immunology, University of Valle, AA 25574 Cali, Colombia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Chappel JA, Rogers WO, Hoffman SL, Kang AS. Molecular dissection of the human antibody response to the structural repeat epitope of Plasmodium falciparum sporozoite from a protected donor. Malar J 2004; 3:28. [PMID: 15283866 PMCID: PMC509279 DOI: 10.1186/1475-2875-3-28] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2004] [Accepted: 07/29/2004] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The circumsporozoite surface protein is the primary target of human antibodies against Plasmodium falciparum sporozoites, these antibodies are predominantly directed to the major repetitive epitope (Asn-Pro-Asn-Ala)n, (NPNA)n. In individuals immunized by the bites of irradiated Anopheles mosquitoes carrying P. falciparum sporozoites in their salivary glands, the anti-repeat response dominates and is thought by many to play a role in protective immunity. METHODS The antibody repertoire from a protected individual immunized by the bites of irradiated P. falciparum infected Anopheles stephensi was recapitulated in a phage display library. Following affinity based selection against (NPNA)3 antibody fragments that recognized the PfCSP repeat epitope were rescued. RESULTS Analysis of selected antibody fragments implied the response was restricted to a single antibody fragment consisting of VH3 and VkappaI families for heavy and light chain respectively with moderate affinity for the ligand. CONCLUSION The dissection of the protective antibody response against the repeat epitope revealed that the response was apparently restricted to a single VH/VL pairing (PfNPNA-1). The affinity for the ligand was in the microM range. If anti-repeat antibodies are involved in the protective immunity elicited by exposure to radiation attenuated P. falciparum sporozoites, then high circulating levels of antibodies against the repeat region may be more important than intrinsic high affinity for protection. The ability to attain and sustain high levels of anti-(NPNA)n will be one of the key determinants of efficacy for a vaccine that relies upon anti-PfCSP repeat antibodies as the primary mechanism of protective immunity against P. falciparum.
Collapse
Affiliation(s)
- Jonathan A Chappel
- Department of Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - William O Rogers
- Malaria Program, Naval Medical Research Center, Silver Spring, MD 20910-7500, USA
- Present address: Naval Medical Research Unit #3, Ghana Det, c/o Department of State, 2020 Accra Place, Washington, DC 20521-2020, USA
| | - Stephen L Hoffman
- Malaria Program, Naval Medical Research Center, Silver Spring, MD 20910-7500, USA
- Present address: Sanaria Inc, 12115 Parklawn Drive Suite L, Rockville, MD 20852, USA
| | - Angray S Kang
- Department of Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
- Present address: Avanir Pharmaceuticals Inc, 11388 Sorrento Valley Road, San Diego, CA 92121, USA
| |
Collapse
|
31
|
Abu-Zeid YA, Alwash R, Shaheen HM, Bin-Othman SA, Lukic ML, Amiri KMA, Charoenvit Y. Seroprevalence of antibodies to repetitive domains of Plasmodium vivax circumsporozoite protein in United Arab Emirates children. Trans R Soc Trop Med Hyg 2002; 96:560-4. [PMID: 12474490 DOI: 10.1016/s0035-9203(02)90443-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The aim of this study was to determine the exposure of child citizens of the United Arab Emirates (UAE) to Plasmodium vivax, and to elucidate if it was related to place of residence or previous international travel to malaria-endemic areas. Blood samples were collected from 1010 primary schoolchildren resident in 7 out of 9 districts of the UAE during October and November 1999. Plasma samples were tested for antibodies against MAP4 (DGQPAGDR)3P2P30, a multiple antigen peptide containing the repeat amino acid sequences of P. vivax circumsporozoite protein (CSP), conjugated to 2 T-helper epitopes, P2 (QYIKANSKFIGITE) and P30 (FNNFTVSFWLRVPKVSASHLE) from tetanus toxin. For confirmation of P. vivax-specific reactivity, positive samples were further tested against (AGDR)6, a synthetic peptide containing 6 copies of a protective epitope within the CSP, and against a recombinant CSP, designated as NS1(81)V20. Results indicated that 3.3% of the children were seropositive. The seropositivity rates differed significantly in relation to place of residence, whereas travel outside the UAE did not significantly affect the exposure rates to P. vivax.
Collapse
Affiliation(s)
- Yousif A Abu-Zeid
- Department of Biology, Faculty of Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.
| | | | | | | | | | | | | |
Collapse
|
32
|
Kumar S, Epstein JE, Richie TL, Nkrumah FK, Soisson L, Carucci DJ, Hoffman SL. A multilateral effort to develop DNA vaccines against falciparum malaria. Trends Parasitol 2002; 18:129-35. [PMID: 11854091 DOI: 10.1016/s1471-4922(01)02207-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Scientists from several organizations worldwide are working together to develop a multistage, multigene DNA-based vaccine against Plasmodium falciparum malaria. This collaborative vaccine development effort is named Multi-Stage DNA-based Malaria Vaccine Operation. An advisory board of international experts in vaccinology, malariology and field trials provides the scientific oversight to support the operation. This article discusses the rationale for the approach, underlying concepts and the pre-clinical development process, and provides a brief outline of the plans for the clinical testing of a multistage, multiantigen malaria vaccine based on DNA plasmid immunization technology.
Collapse
Affiliation(s)
- Sanjai Kumar
- Malaria Program, Naval Medical Research Center, Silver Spring, MD 20910, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Plasmodium vivax represents the most widespread malaria parasite worldwide. Although it does not result in as high a mortality rate as P. falciparum, it inflicts debilitating morbidity and consequent economic impact in endemic communities. In addition, the relapsing behavior of this malaria parasite and the recent resistance to anti-malarials contribute to making its control more difficult. Although the biology of P. vivax is different from that of P. falciparum and the human immune response to this parasite species has been rather poorly studied, significant progress is being made to develop a P. vivax-specific vaccine based on the information and experience gained in the search for a P. falciparum vaccine. We have devoted great effort to antigenically characterize the P. vivax CS protein and to test its immunogenicity using the Aotus monkey model. Together with other groups we are also assessing the immunogenicity and protective efficacy of the asexual blood stage vaccine candidates MSP-1 and DBP in the monkey model, as well as the immunogenicity of Pvs25 and Pvs28 ookinete surface proteins. The transmission-blocking efficacy of the responses induced by these latter antigens is being assessed using Anopheles albimanus mosquitoes. The current status of these vaccine candidates and other antigens currently being studied is described.
Collapse
|
34
|
Brahimi K, Badell E, Sauzet JP, BenMohamed L, Daubersies P, Guérin-Marchand C, Snounou G, Druilhe P. Human antibodies against Plasmodium falciparum liver-stage antigen 3 cross-react with Plasmodium yoelii preerythrocytic-stage epitopes and inhibit sporozoite invasion in vitro and in vivo. Infect Immun 2001; 69:3845-52. [PMID: 11349050 PMCID: PMC98406 DOI: 10.1128/iai.69.6.3845-3952.2001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Plasmodium falciparum liver-stage antigen 3 (LSA3), a recently identified preerythrocytic antigen, induces protection against malaria in chimpanzees. Using antibodies from individuals with hyperimmunity to malaria affinity purified on recombinant or synthetic polypeptides of LSA3, we identified four non-cross-reactive B-cell epitopes in Plasmodium yoelii preerythrocytic stages. On sporozoites the P. yoelii protein detected has a molecular mass similar to that of LSA3. T-cell epitopes cross-reacting with P. yoelii were also demonstrated using peripheral blood lymphocytes from LSA3-immunized chimpanzees. In contrast, no cross-reactive epitopes were found in Plasmodium berghei. LSA3-specific human antibodies exerted up to 100% inhibition of in vitro invasion of P. yoelii sporozoites into mouse hepatocytes. This strong in vitro activity was reproduced in vivo by passive transfer of LSA3 antibodies. These results indicate that the homologous epitopes may be biologically functional and suggest that P. yoelii could be used as a model to assess the antisporozoite activity of anti-LSA3 antibodies.
Collapse
Affiliation(s)
- K Brahimi
- Laboratoire de Parasitologie Biomédicale, Institut Pasteur, 75015 Paris Cedex 15, France
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Charoenvit Y, Majam VF, Corradin G, Sacci JB, Wang R, Doolan DL, Jones TR, Abot E, Patarroyo ME, Guzman F, Hoffman SL. CD4(+) T-cell- and gamma interferon-dependent protection against murine malaria by immunization with linear synthetic peptides from a Plasmodium yoelii 17-kilodalton hepatocyte erythrocyte protein. Infect Immun 1999; 67:5604-14. [PMID: 10531206 PMCID: PMC96932 DOI: 10.1128/iai.67.11.5604-5614.1999] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Most work on protective immunity against the pre-erythrocytic stages of malaria has focused on induction of antibodies that prevent sporozoite invasion of hepatocytes, and CD8(+) T-cell responses that eliminate infected hepatocytes. We recently reported that immunization of A/J mice with an 18-amino-acid synthetic linear peptide from Plasmodium yoelii sporozoite surface protein 2 (SSP2) in TiterMax adjuvant induces sterile protection that is dependent on CD4(+) T cells and gamma interferon (IFN-gamma). We now report that immunization of inbred A/J mice and outbred CD1 mice with each of two linear synthetic peptides from the 17-kDa P. yoelii hepatocyte erythrocyte protein (HEP17) in the same adjuvant also induces protection against sporozoite challenge that is dependent on CD4(+) T cells and IFN-gamma. The SSP2 peptide and the two HEP17 peptides are recognized by B cells as well as T cells, and the protection induced by these peptides appears to be directed against the infected hepatocytes. In contrast to the peptide-induced protection, immunization of eight different strains of mice with radiation-attenuated sporozoites induces protection that is absolutely dependent on CD8(+) T cells. Data represented here demonstrate that CD4(+) T-cell-dependent protection can be induced by immunization with linear synthetic peptides. These studies therefore provide the foundation for an approach to pre-erythrocytic-stage malaria vaccine development, based on the induction of protective CD4(+) T-cell responses, which will complement efforts to induce protective antibody and CD8(+) T-cell responses.
Collapse
Affiliation(s)
- Y Charoenvit
- Malaria Program, Naval Medical Research Center, Bethesda, Maryland 20814-5055, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Rogers WO, Gowda K, Hoffman SL. Construction and immunogenicity of DNA vaccine plasmids encoding four Plasmodium vivax candidate vaccine antigens. Vaccine 1999; 17:3136-44. [PMID: 10462250 DOI: 10.1016/s0264-410x(99)00146-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Plasmodium vivax is the second most common agent of human malaria. Although infection is rarely fatal, it nonetheless imposes a significant burden of illness in endemic areas. A successful vaccine against P. vivax will likely need to induce immune responses against both pre-erythrocytic and erythrocytic stage forms of the parasite. Accordingly, we constructed eight nucleic acid vaccines based on four antigens, the circumsporozoite protein (PvCSP) and sporozoite surface protein 2 (PvSSP2) from the pre-erythrocytic stage, and apical membrane antigen 1 (PvAMA1) and merozoite surface protein 1 (PvMSP1) from the erythrocytic stage. The constructs induced high levels of specific antibody in mice regardless of whether the antigen was expressed in native form or fused to a human tissue plasminogen activator leader peptide. High titer antibodies induced against PvCSP did not react with the protective AGDR epitope within the sequence of this antigen. These results support the immunogenicity of these four vaccine candidate antigens when delivered as nucleic acid vaccines.
Collapse
Affiliation(s)
- W O Rogers
- Naval Medical Research Center, Bethesda, MD, USA.
| | | | | |
Collapse
|
37
|
Le TP, Church LW, Corradin G, Hunter RL, Charoenvit Y, Wang R, de la Vega P, Sacci J, Ballou WR, Kolodny N, Kitov S, Glenn GM, Richards RL, Alving CR, Hoffman SL. Immunogenicity of Plasmodium falciparum circumsporozoite protein multiple antigen peptide vaccine formulated with different adjuvants. Vaccine 1998; 16:305-12. [PMID: 9607047 DOI: 10.1016/s0264-410x(97)00165-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Only low antibody levels were obtained from vaccinating human volunteers with single-chain peptide from the Plasmodium falciparum circumsporozoite protein (PfCSP). This resulted in modest protection against sporozoite challenge. In addition, HLA restriction limits the probability of synthesis of a vaccine effective for a diverse population. We report immunization studies with a multiple antigen peptide (MAP) system consisting of multiple copies of a B-cell epitope from the central repeat region of the PfCSP in combination with a universal T-cell epitope, the P2P30 portion of tetanus toxin. This MAP4(NANP)6P2P30 vaccine was highly immunogenic in four different strains of mice when used with various safe and nontoxic adjuvants. When this MAP vaccine was encapsulated in liposomes with lipid A and adsorbed to aluminium hydroxide and given three times at 4-week intervals, the resultant antibody prevented 100% of sporozoites from invading and developing into liver stage infection. This high degree of immunogenicity of MAP4(NANP)6P2P30 vaccine formulated in liposomes, lipid A and aluminum hydroxide provides the foundation for consideration of human trials with this formulation.
Collapse
Affiliation(s)
- T P Le
- Malaria Program, Naval Medical Research Institute, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Doolan DL, Hoffman SL. Pre-erythrocytic-stage immune effector mechanisms in Plasmodium spp. infections. Philos Trans R Soc Lond B Biol Sci 1997; 352:1361-7. [PMID: 9355128 PMCID: PMC1692030 DOI: 10.1098/rstb.1997.0121] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The potent protective immunity against malaria induced by immunization of mice and humans with radiation-attenuated Plasmodium spp. sporozoites is thought to be mediated primarily by T-cell responses directed against infected hepatocytes. This has led to considerable efforts to develop subunit vaccines that duplicate this protective immunity, but a universally effective vaccine is still not available and in vitro correlates of protective immunity have not been established. Contributing to this delay has been a lack of understanding of the mechanisms responsible for the protection. There are now data indicating that CD8+ T cells, CD4+ T cells, cytokines, and nitric oxide can all mediate the elimination of infected hepatocytes in vitro and in vivo. By dissecting the protection induced by immunization with irradiated sporozoite, DNA and synthetic peptide-adjuvant vaccines, we have demonstrated that different T-cell-dependent immune responses mediate protective immunity in the same inbred strain of mouse, depending on the method of immunization. Furthermore, the mechanism of protection induced by a single method of immunization may vary among different strains of mice. These data have important implications for the development of pre-erythrocytic-stage vaccines designed to protect a heterogeneous human population, and of assays that predict protective immunity.
Collapse
Affiliation(s)
- D L Doolan
- Malaria Program, Naval Medical Research Institute, Rockville, MD 20852, USA
| | | |
Collapse
|
39
|
Charoenvit Y, Fallarme V, Rogers WO, Sacci JB, Kaur M, Aguiar JC, Yuan LF, Corradin G, Andersen E, Wizel B, Houghten RA, Oloo A, De la Vega P, Hoffman SL. Development of two monoclonal antibodies against Plasmodium falciparum sporozoite surface protein 2 and mapping of B-cell epitopes. Infect Immun 1997; 65:3430-7. [PMID: 9234808 PMCID: PMC175485 DOI: 10.1128/iai.65.8.3430-3437.1997] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The Plasmodium yoelii sporozoite surface protein 2 (PySSP2) is the target of protective cellular immunity. Cytotoxic T cells specific for the Plasmodium falciparum analog PfSSP2, also known as thrombospondin-related anonymous protein (TRAP), are induced in human volunteers immunized with irradiated sporozoites. PfSSP2 is an important candidate antigen for a multicomponent malaria vaccine. We generated and characterized three monoclonal antibodies (MAbs) specific for PfSSP2/TRAP. The MAbs PfSSP2.1 (immunoglobulin G1 [IgG1]), PfSSP2.2 (IgG2a), and PfSSP2.3 (IgM) were species specific and identified three distinct B-cell epitopes containing sequences DRYI, CHPSDGKC, and TRPHGR, respectively. PfSSP2.1 partially inhibited P. falciparum liver-stage parasite development in human hepatocyte cultures (42 and 86% in two experiments at 100 microg/ml). Mice immunized with vaccinia virus expressing full-length PfSSP2 protein produced antibodies to (DRYIPYSP)3, and humans living in malaria-endemic areas (Indonesia and Kenya), who have lifelong exposure and partial clinical immunity to malaria, had antibodies to both (DRYIPYSP)3 and (CHPSDGKCN)2. Mice immunized with multiple antigen peptides MAP4 (DRYIPYSP)3P2P30 and MAP4 (CHPSDGKCN)3P2P30 in TiterMax developed antibodies to sporozoites that partially inhibited sporozoite invasion of human hepatoma cells (39 to 71% at a serum dilution of 1:50 in three different experiments). The modest inhibitory activities of the MAbs and the polyclonal antibodies to PfSSP2/TRAP epitopes do not suggest that a single-component vaccine designed to induce antibodies against PfSSP2/TRAP will be protective. Nonetheless, the MAbs directed against PfSSP2, and the peptides recognized by these MAbs, will be essential reagents in the development of PfSSP2/TRAP as a component of a multivalent P. falciparum human malaria vaccine.
Collapse
Affiliation(s)
- Y Charoenvit
- Malaria Program, Naval Medical Research Institute, Bethesda, Maryland 20889-5607, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Bilsborough J, Baumgart K, Bathurst I, Barr P, Good MF. Fine epitope specificity of antibodies to region II of the Plasmodium vivax circumsporozoite protein correlates with ability to bind recombinant protein and sporozoites. Acta Trop 1997; 65:59-80. [PMID: 9164601 DOI: 10.1016/s0001-706x(97)00648-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Recent work has suggested that important B- and T-cell epitopes on the circumsporozoite protein (CSP) of Plasmodium vivax lie external to the major repeat regions of the protein. We have studied two naturally exposed human populations (Caucasian and Papua New Guineans) and determined the antibody response to yeast-derived recombinant CSPs, overlapping synthetic peptides spanning amino acids 76 348 of the Belem P. vivax CSP and overlapping peptides representing the variant repeats of the VK247 strain of P. vivax. We have demonstrated that the P. vivax CSP-specific antibody response is directed towards areas within the repeat region as well as areas external to this; but the dominant epitopes recognized by the two populations studied, were distinct. One epitope, lying external to the repeats and recognized by both populations, partially overlaps an area of the protein referred to as region II-plus. Sera from malaria-exposed Papua New Guineans and Thais contained antibodies to this epitope (V22, single letter amino acid sequence TCGVGVRVRRRVNAANKKPE) which were capable of recognizing sporozoites, as determined by quantitative inhibition IFA. Seventeen percent of PNG sera had antibodies to this peptide compared with 33% who had antibodies to the central repeats of the protein. Immunization of mice with recombinant CSP did not induce antibodies to V22. However, immunization with overlapping peptide epitopes representing this region (V21 or V22) induced specific antibodies but only two sera recognized both V21 and V22 and, by inference, the overlapping peptide sequence (TCGVGVRVRR). Antibodies in these two sera could bind recombinant CSP in ELISA; however, in contrast, nine sera which recognized either V21 or V22 alone did not bind CSP. Only one of two sera containing antibodies recognizing CSP stained P. vivax sporozoites. This serum also recognized an epitope dependent upon two amino acids aminoterminal to V22. These data suggest that the fine specificity of antibodies is a critical determinant for binding to both recCSP and sporozoites.
Collapse
Affiliation(s)
- J Bilsborough
- Queensland Institute of Medical Research, Royal Brisbane Hospital, Brisbane, Australia
| | | | | | | | | |
Collapse
|
41
|
Doolan DL, Hoffman SL. Multi-gene vaccination against malaria: A multistage, multi-immune response approach. ACTA ACUST UNITED AC 1997; 13:171-8. [PMID: 15275087 DOI: 10.1016/s0169-4758(97)01040-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
An ideal malaria vaccine will induce immune responses against each stage of the Plasmodium spp life cycle. During its complicated life cycle, the parasite exists extracellularly in the host's bloodstream, within cells that express major histocompatibility complex (MHC) molecules (hepatocytes), within cells that do not express MHC molecules (erythrocytes) and within the mosquito vector. Different arms of the immune system are required to attack the parasite at the different stages. Therefore, a multistage vaccine must be a multi-immune response vaccine. In addition, given the unique antigenicities of the different stages of the life cycle, implicit in this definition is that the vaccine be multivalent. Here, Denise Doolan and Stephen Hoffman present the rationale for developing a multistage, multivalent, multi-immune response malaria vaccine and explain why, among currently available technologies, DNA vaccines may offer the best prospect for success.
Collapse
Affiliation(s)
- D L Doolan
- Malaria Program, Naval Medical Research Institute, 12300 Washington Avenue, Rockville, MD 20852, USA. doolan@nmripo/.nmri.nnmc.navy.mil
| | | |
Collapse
|
42
|
Reed RC, Louis-Wileman V, Cosmai EV, Fang S, Jue DL, Wohlhueter RM, Hunter RL, Lal AA. Multiple antigen constructs (MACs): induction of sterile immunity against sporozoite stage of rodent malaria parasites, Plasmodium berghei and Plasmodium yoelii. Vaccine 1997; 15:482-8. [PMID: 9160515 DOI: 10.1016/s0264-410x(96)00301-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We prepared multiple antigen constructs (MACs) using circumsporozoite (CS) protein-based B-epitopes from Plasmodium berghei, (PPPPNPND)2 and Plasmodium yoelii, (QGPGAP)3QG, along with a P. berghei T-helper epitope KQIRDSITEEWS. Mice were immunized with individual MACs in oil-in-water or water-in-oil vehicles containing block copolymer (P1005) and detoxified RaLPS (RaLPS) as well as other adjuvants. Sporozoite challenge results demonstrated that MACs in adjuvant could induce antibodies capable of active and passive protection. Water-in-oil vaccines induced the highest level of protection in mice immunized with either P. berghei or P. yoelii MACs. In a study aimed at co-eliciting immunity against P. berghei and P. yoelii, three immunizations with MACs induced protective antibodies against P. berghei but not P. yoelii parasite challenge. Therefore, it can be concluded that individually MACs are capable of inducing strong and protective immune responses to either species of rodent malaria, and that protection can be passively transferred. When MAC formulations were used together as a combined vaccine, P. berghei MACs induced a strong protective antibody response while P. yoelii MACs induced a weaker nonprotective response.
Collapse
Affiliation(s)
- R C Reed
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Yang C, Collins WE, Xiao L, Saekhou AM, Reed RC, Nelson CO, Hunter RL, Jue DL, Fang S, Wohlhueter RM, Udhayakumar V, Lal AA. Induction of protective antibodies in Saimiri monkeys by immunization with a multiple antigen construct (MAC) containing the Plasmodium vivax circumsporozoite protein repeat region and a universal T helper epitope of tetanus toxin. Vaccine 1997; 15:377-86. [PMID: 9141208 DOI: 10.1016/s0264-410x(97)00200-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Previous attempts in inducing protective immunity against Plasmodium vivax in human volunteers and nonhuman primates with recombinant circumsporozoite (CS) proteins have been unsuccessful, largely due to the failure of generating antibodies against the protective B epitope AGDR in the CS protein repeat region. We report here an immunization study in Saimiri monkeys with a multiple antigen construct (MAC) containing the P. vivax CS protein repeat region and a T helper epitope of tetanus toxin formulated in different adjuvants. Monkeys immunized three times with MAC in copolymer P1005, copolymer P1005 plus RaLPS, or MF-75 had titers of antibodies against CS repeat, sporozoites and the protective B epitope AGDR significantly higher than those immunized with MAC in alum or PBS (P < 0.05). Antibody levels in animals that received P1005 were maintained at high level for 7 months after the last immunization. Upon challenge with 10000 sporozoites 2 weeks after the last immunization, 75% (three of four) of monkeys from the alum group, 50% (three of six) of monkeys from the P1005 plus RaLPS group, 40% (two of five) of monkeys from the P1005 group, 33% (two of six) of monkeys from the MF-75 group, and 17% (one of six) of monkeys from the MAC alone group were fully protected. When immunized animals were challenged again with 30000 sporozoites 22 weeks after the last immunization. 40% (two of five) monkeys from the P1005 group were fully protected. The remaining (three) in this group developed low parasitemia (< 2000 parasites mm-3 of blood) after significantly longer prepatent period (P < 0.05). In addition, 17% (one of six) of monkeys each from the P1005 plus RaLPS and MF-75 groups were also fully protected. Protected animals had higher levels of prechallenge anti-AGDR antibody titers than unprotected (1933 vs 281 for the first challenge, P > 0.05; 21527 vs 196 for the rechallenge, P < 0.05). Anti-AGDR antibody titers were positively correlated with the prepatent period of infected animals (r = 0.42 for the first challenge, P > 0.05; r = 0.60 for the rechallenge, P < 0.05) and negatively correlated with the peak parasitemia (r = -0.39 for the first challenge, P < 0.05; r = 0.50 for the rechallenge, P < 0.05). The results suggested that when combined with the use of potent adjuvants and T helper epitopes, MAC subunit vaccines may potentially offer protection against malaria infection.
Collapse
Affiliation(s)
- C Yang
- Division of Parasitic Diseases, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ahlborg N, Iqbal J, Hansson M, Uhlén M, Mattei D, Perlmann P, Stahl S, Berzins K. Immunogens containing sequences from antigen Pf332 induce Plasmodium falciparum-reactive antibodies which inhibit parasite growth but not cytoadherence. Parasite Immunol 1995; 17:341-52. [PMID: 8552406 DOI: 10.1111/j.1365-3024.1995.tb00901.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Immunogens based upon sequences from the P. falciparum asexual blood stage antigen Pf332 were assessed for their capacity to induce antibodies inhibiting parasite growth or cytoadherence of infected erythrocytes in vitro. Selection of the Pf332 sequences was based on their reactivity with the human monoclonal antibody (MoAb) 33G2 which inhibits parasite growth as well as cytoadherence in vitro. Octameric multiple antigen peptides (MAP) were assembled based upon either a trimer of the minimal epitope recognized by the MoAb, VTEEI, or a Pf332 sequence including that motif, SVTEEIAEEDK. A dimer of SVTEEIAEEDK was also expressed in Escherichia coli, genetically fused to ZZ, two IgG-binding domains of staphylococcal protein A. Rabbit antibodies elicited by the immunogens reacted with Pf332 in immunofluorescence and in ELISA with Pf332 peptides which were also recognized by MoAb 33G2. The MAP with branched (VTEEI)3 peptide induced the highest titres of P. falciparum-reactive antibodies. In contrast to MoAB 33G2, none of the polyclonal Pf332 reactive sera cross-reacted with repeat sequences of the malaria antigen Pf155/RESA. The polyclonal Pf332-reactive antibodies inhibited parasite growth efficiently but had no or very low inhibitory effect in a cytoadherence assay. Thus, while Pf332 may be an important target for parasite neutralizing antibodies its involvement in cytoadherence is unclear.
Collapse
Affiliation(s)
- N Ahlborg
- Department of Immunology, Stockholm University, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Etlinger HM. Selection of carrier and B cell protectope sequences for vaccines. Ann N Y Acad Sci 1995; 754:57-67. [PMID: 7625681 DOI: 10.1111/j.1749-6632.1995.tb44438.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- H M Etlinger
- F. Hoffmann-La Roche & Company, Basel, Switzerland
| |
Collapse
|
46
|
Turpen TH, Reinl SJ, Charoenvit Y, Hoffman SL, Fallarme V, Grill LK. Malarial epitopes expressed on the surface of recombinant tobacco mosaic virus. Nat Biotechnol 1995; 13:53-7. [PMID: 9634749 DOI: 10.1038/nbt0195-53] [Citation(s) in RCA: 174] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Using malaria as a model disease, we engineered the surface of tobacco mosaic tobamovirus (TMV) for presentation of selected epitopes to the mammalian immune system. The TMV coat protein is a well-characterized and abundant self-assembling polymer previously shown to be a highly immunogenic carrier. Selected B-cell epitopes were either inserted into the surface loop region of the TMV coat protein or fused to the C terminus using the leaky stop signal derived from the replicase protein reading frame. Tobacco plants systemically infected with each of these constructs contained high titers of genetically stable recombinant virus, enabling purification of the chimeric particles in high yield. Symptoms induced in tobacco ranged from a normal mosaic pattern similar to that induced by the parental U1 strain to a unique bright yellow mosaic. As measured by quantitative ELISA against synthetic peptide standards, wild type TMV coat protein and fusion protein synthesized by the leaky stop mechanism coassembled into virus particles at the predicted ratio of approximately 20:1. Recombinant plant viruses have the potential to meet the need for scalable and cost effective production of subunit vaccines that can be easily stored and administered.
Collapse
Affiliation(s)
- T H Turpen
- Biosource Technologies, Inc., Vacaville, CA 95688.
| | | | | | | | | | | |
Collapse
|
47
|
Hoffman SL, Sacci JB. Rationale and approaches to constructing preerythrocytic malaria vaccines. PHARMACEUTICAL BIOTECHNOLOGY 1995; 6:787-802. [PMID: 7551248 DOI: 10.1007/978-1-4615-1823-5_35] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- S L Hoffman
- Malaria Program, Naval Medical Research Institute, Bethesda, Maryland 20889-5607, USA
| | | |
Collapse
|
48
|
Mann V, Good M, Saul A. Diversity in the circumsporozoite protein of Plasmodium vivax: Does it matter? ACTA ACUST UNITED AC 1995. [DOI: 10.1016/0169-4758(95)80107-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
49
|
Hoffman SL, Sedegah M, Hedstrom RC. Protection against malaria by immunization with a Plasmodium yoelii circumsporozoite protein nucleic acid vaccine. Vaccine 1994; 12:1529-33. [PMID: 7879419 DOI: 10.1016/0264-410x(94)90078-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Nucleic acid vaccines provide an exciting new alternative approach to developing the multiantigen vaccines designed to induce protective antibody and T-cell responses against Plasmodium proteins that many experts believe will be required for effective protection against malaria. As a first step in this process, we produced a plasmid DNA vaccine that includes the gene encoding the P. yoelii circumsporozoite protein (PyCSP). This vaccine induced higher levels of antibodies and cytotoxic T lymphocytes against PyCSP than immunization with irradiated sporozoites, and protected 9 of the first 16 mice immunized. Work is now in progress to optimize immunization regimens, establish the mechanisms of protective immunity induced by the vaccine, and to determine whether protective immunity can be increased by vaccinating with multiple nucleic acid vaccines designed to produce immune responses against multiple targets.
Collapse
Affiliation(s)
- S L Hoffman
- Malaria Program, Naval Medical Research Institute, Bethesda, MD 20889-5607
| | | | | |
Collapse
|
50
|
Schödel F, Wirtz R, Peterson D, Hughes J, Warren R, Sadoff J, Milich D. Immunity to malaria elicited by hybrid hepatitis B virus core particles carrying circumsporozoite protein epitopes. J Exp Med 1994; 180:1037-46. [PMID: 7520465 PMCID: PMC2191626 DOI: 10.1084/jem.180.3.1037] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The hepatitis B virus (HBV) nucleocapsid antigen (HBcAg) was investigated as a carrier moiety for the immunodominant circumsporozoite (CS) protein repeat epitopes of Plasmodium falciparum and the rodent malaria agent P. berghei. For this purpose hybrid genes coding for [NANP]4 (C75CS2) or [DP4NPN]2 (C75CS1) as internal inserts in HBcAg (between amino acids 75 and 81) were constructed and expressed in recombinant Salmonella typhimurium. The resulting hybrid HBcAg-CS polypeptides purified from S. typhimurium were particulate and displayed CS and HBc antigenicity, however, the HBc antigenicity was reduced compared to native recombinant HBcAg. Immunization of several mouse strains with HBcAg-CS1 and HBcAg-CS2 particles resulted in high titer, P.berghei- or P.falciparum-specific anti-CS antibodies representing all murine immunoglobulin G isotypes. The possible influence of carrier-specific immunosuppression was examined, and preexisting immunity to HBcAg did not significantly affect the immunogenicity of the CS epitopes within HBcAg-CS1 particles. Similarly, the choice of adjuvant did not significantly alter the immunogenicity of HBcAg-CS hybrid particles. Immunization in complete or incomplete Freund's adjuvant or alum resulted in equivalent anti-HBc and anti-CS humoral responses. Examination of T cell recognition of HBcAg-CS particles revealed that HBcAg-specific T cells were universally primed and CS-specific T cells were primed if the insert contained a CS-specific T cell recognition site. This indicates that the internal site in HBcAg is permissive for the inclusion of heterologous pathogen-specific T as well as B cell epitopes. Most importantly, 90 and 100% of BALB/c mice immunized with HBcAg-CS1 particles were protected against a P. berghei challenge infection in two independent experiments. Therefore, hybrid HBcAg-CS particles may represent a useful approach for future malaria vaccine development.
Collapse
Affiliation(s)
- F Schödel
- Department of Bacterial Diseases, Walter Reed Army Institute of Research, Washington, DC 20307-5100
| | | | | | | | | | | | | |
Collapse
|