1
|
Horn M, Bieliková L, Vostoupalová A, Švéda J, Mareš M. An update on proteases and protease inhibitors from trematodes. ADVANCES IN PARASITOLOGY 2024; 126:97-176. [PMID: 39448195 DOI: 10.1016/bs.apar.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Trematodes, a class of parasitic flatworms, are responsible for a variety of devastating diseases in humans and animals, with schistosomiasis and fascioliasis being prominent examples. Trematode proteolytic systems involved in the host-parasite interaction have emerged as key contributors to the success of trematodes in establishing and maintaining infections. This review concentrates on diverse proteases and protease inhibitors employed by trematodes and provides an update on recent advances in their molecular-level characterization, with a focus on function, structure, and therapeutic target potential.
Collapse
Affiliation(s)
- Martin Horn
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Lucia Bieliková
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Andrea Vostoupalová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jakub Švéda
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Michael Mareš
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
2
|
Ekloh W, Asafu-Adjaye A, Tawiah-Mensah CNL, Ayivi-Tosuh SM, Quartey NKA, Aiduenu AF, Gayi BK, Koudonu JAM, Basing LA, Yamoah JAA, Dofuor AK, Osei JHN. A comprehensive exploration of schistosomiasis: Global impact, molecular characterization, drug discovery, artificial intelligence and future prospects. Heliyon 2024; 10:e33070. [PMID: 38988508 PMCID: PMC11234110 DOI: 10.1016/j.heliyon.2024.e33070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024] Open
Abstract
Schistosomiasis, one of the neglected tropical diseases which affects both humans and animals, is caused by trematode worms of the genus Schistosoma. The disease is caused by several species of Schistosoma which affect several organs such as urethra, liver, bladder, intestines, skin and bile ducts. The life cycle of the disease involves an intermediate host (snail) and a mammalian host. It affects people who are in close proximity to water bodies where the intermediate host is abundant. Common clinical manifestations of the disease at various stages include fever, chills, headache, cough, dysuria, hyperplasia and hydronephrosis. To date, most of the control strategies are dependent on effective diagnosis, chemotherapy and public health education on the biology of the vectors and parasites. Microscopy (Kato-Katz) is considered the golden standard for the detection of the parasite, while praziquantel is the drug of choice for the mass treatment of the disease since no vaccines have yet been developed. Most of the previous reviews on schistosomiasis have concentrated on epidemiology, life cycle, diagnosis, control and treatment. Thus, a comprehensive review that is in tune with modern developments is needed. Here, we extend this domain to cover historical perspectives, global impact, symptoms and detection, biochemical and molecular characterization, gene therapy, current drugs and vaccine status. We also discuss the prospects of using plants as potential and alternative sources of novel anti-schistosomal agents. Furthermore, we highlight advanced molecular techniques, imaging and artificial intelligence that may be useful in the future detection and treatment of the disease. Overall, the proper detection of schistosomiasis using state-of-the-art tools and techniques, as well as development of vaccines or new anti-schistosomal drugs may aid in the elimination of the disease.
Collapse
Affiliation(s)
- William Ekloh
- Department of Biochemistry, School of Biological Sciences, College of Agriculture and Natural Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Andy Asafu-Adjaye
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Christopher Nii Laryea Tawiah-Mensah
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | | | - Naa Kwarley-Aba Quartey
- Department of Food Science and Technology, Faculty of Biosciences, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Albert Fynn Aiduenu
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra, Ghana
| | - Blessing Kwabena Gayi
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra, Ghana
| | | | - Laud Anthony Basing
- Department of Medical Diagnostics, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Jennifer Afua Afrifa Yamoah
- Animal Health Division, Council for Scientific and Industrial Research-Animal Research Institute, Adenta-Frafraha, Accra, Ghana
| | - Aboagye Kwarteng Dofuor
- Department of Biological Sciences, School of Natural and Environmental Sciences, University of Environment and Sustainable Development, Somanya, Ghana
| | - Joseph Harold Nyarko Osei
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| |
Collapse
|
3
|
Allen T, Castellanos ME, Giacomin P, Karunaweera ND, Kupz A, Lol JC, Sharma D, Sikder S, Tedla B, van Eijk L, Vojisavljevic D, Zhao G, Pai S. Next-generation vaccines for tropical infectious diseases. Int J Infect Dis 2024; 143:107014. [PMID: 38499058 DOI: 10.1016/j.ijid.2024.107014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/05/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024] Open
Abstract
Tropical infectious diseases inflict an unacceptable burden of disease on humans living in developing countries. Although anti-pathogenic drugs have been widely used, they carry a constant threat of selecting for resistance. Vaccines offer a promising means by which to enhance the global control of tropical infectious diseases; however, these have been difficult to develop, mostly because of the complex nature of the pathogen lifecycles. Here, we present recently developed vaccine candidates for five tropical infectious diseases in the form of a catalog that have either entered clinical trials or have been licensed for use. We deliberate on recently licensed dengue vaccines, provide evidence why combination vaccination could have a synergistic impact on schistosomiasis, critically appraise the value of typhoid conjugate vaccines, and discuss the potential of vaccines in the efforts to eliminate vivax malaria and hookworms.
Collapse
Affiliation(s)
- Tammy Allen
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia
| | - Maria Eugenia Castellanos
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia; Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Paul Giacomin
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | | | - Andreas Kupz
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Juan Carlos Lol
- Centro de Estudios en Salud, Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | - Dileep Sharma
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia; College of Medicine & Dentistry, James Cook University, Cairns, Australia; School of Health Sciences, College of Health Medicine and Wellbeing, The University of Newcastle, Ourimbah, Australia
| | - Suchandan Sikder
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Bemnet Tedla
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia; Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Liza van Eijk
- Department of Psychology, James Cook University, Townsville, Australia
| | - Danica Vojisavljevic
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia; Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Guangzu Zhao
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Saparna Pai
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia.
| |
Collapse
|
4
|
Elguindy DAS, Ashour DS, Elmarhoumy SM, El-Guindy DM, Ismail HIH. The efficacy of cercarial antigen loaded on nanoparticles as a potential vaccine candidate in Schistosoma mansoni-infected mice. J Parasit Dis 2024; 48:381-399. [PMID: 38840868 PMCID: PMC11147980 DOI: 10.1007/s12639-024-01677-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 04/21/2024] [Indexed: 06/07/2024] Open
Abstract
Schistosomiasis is one of the most common causes of morbidity and mortality from parasitic diseases. Mass treatment has proven to be insufficient because of repeated infection after treatment and the appearance of strains resistant to drug therapy. Hence, immunization is a new approach to control the disease and limit the pathological consequences of schistosomiasis. To evaluate the prophylactic effect of Cercarial antigen (CAP) loaded on chitosan nanoparticles (CSNPs) as a potential vaccine against Schistosoma mansoni-infected mice. 130 mice divided into 2 groups were used: Group I: Control groups (50 mice) subdivided into subgroup Ia (10 mice): Non-infected mice (normal control), subgroup Ib (20 mice): Schistosoma infected mice (infected control) and subgroup Ic (20 mice): Non-infected mice receiving NPs only. Group II: Vaccinated group (80 mice) subdivided equally into subgroup IIa (CAP): Received cercarial antigen and subgroup IIb (CAP + CSNP): Received cercarial antigen loaded on chitosan NPs then both vaccinated groups were infected with S. mansoni 3 weeks following the initial vaccination dose. CAP + CSNP and CAP groups showed significant reduction in adult worms count, hepatic egg count, hepatic granulomas number and size in comparison to the infected control group. Elevation of serum IgG and IgM levels, CD4+ and CD8+ T cell frequencies, IL-4, IL-10 and INF-γ levels was more significant in CAP + CSNP group than CAP group. CAP + CSNP is a promising new preparation of Schistosomal antigens that gave better results than immunization with CAP alone. CSNPs enhanced the immune and protective effect of CAP as validated by parasitological, histopathological and immunohistochemical studies.
Collapse
Affiliation(s)
- Dina A. S. Elguindy
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Dalia S. Ashour
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Sirria M. Elmarhoumy
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Dina M. El-Guindy
- Pathology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Howaida I. H. Ismail
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
5
|
Barban do Patrocinio A. Schistosomiasis: Discovery of New Molecules for Disease Treatment and Vaccine Development. Infect Dis (Lond) 2022. [DOI: 10.5772/intechopen.104738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The parasite blood flukes belonging to the genus Schistosoma cause schistosomiasis. Among the Schistosoma species that infect humans, three stand out: Schistosoma japonicum (S. japonicum), which occurs in Asia, mainly in China and the Philippines; Schistosoma haematobium (S. haematobium), which occurs in Africa; and Schistosoma mansoni (S. mansoni), which occurs in Africa and South America and the center of Venezuela (Brazil). Research has shown that these species comprise strains that are resistant to Praziquantel (PZQ), the only drug of choice to fight the disease. Moreover, patients can be reinfected even after being treated with PZQ , and this drug does not act against young forms of the parasite. Therefore, several research groups have focused their studies on new molecules for disease treatment and vaccine development. This chapter will focus on (i) parasite resistance to PZQ , (ii) molecules that are currently being developed and tested as possible drugs against schistosomiasis, and (iii) candidates for vaccine development with a primary focus on clinical trials.
Collapse
|
6
|
Vaccines for Human Schistosomiasis: Recent Progress, New Developments and Future Prospects. Int J Mol Sci 2022; 23:ijms23042255. [PMID: 35216369 PMCID: PMC8879820 DOI: 10.3390/ijms23042255] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 12/18/2022] Open
Abstract
Schistosomiasis, caused by human trematode blood flukes (schistosomes), remains one of the most prevalent and serious of the neglected tropical parasitic diseases. Currently, treatment of schistosomiasis relies solely on a single drug, the anthelmintic praziquantel, and with increased usage in mass drug administration control programs for the disease, the specter of drug resistance developing is a constant threat. Vaccination is recognized as one of the most sustainable options for the control of any pathogen, but despite the discovery and reporting of numerous potentially promising schistosome vaccine antigens, to date, no schistosomiasis vaccine for human or animal deployment is available. This is despite the fact that Science ranked such an intervention as one of the top 10 vaccines that need to be urgently developed to improve public health globally. This review summarizes current progress of schistosomiasis vaccines under clinical development and advocates the urgent need for the establishment of a revolutionary and effective anti-schistosome vaccine pipeline utilizing cutting-edge technologies (including developing mRNA vaccines and exploiting CRISPR-based technologies) to provide novel insight into future vaccine discovery, design, manufacture and deployment.
Collapse
|
7
|
Panzner U, Excler JL, Kim JH, Marks F, Carter D, Siddiqui AA. Recent Advances and Methodological Considerations on Vaccine Candidates for Human Schistosomiasis. FRONTIERS IN TROPICAL DISEASES 2021; 2:719369. [PMID: 39280170 PMCID: PMC11392908 DOI: 10.3389/fitd.2021.719369] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2024] Open
Abstract
Schistosomiasis remains a neglected tropical disease of major public health concern with high levels of morbidity in various parts of the world. Although considerable efforts in implementing mass drug administration programs utilizing praziquantel have been deployed, schistosomiasis is still not contained. A vaccine may therefore be an essential part of multifaceted prevention control efforts. In the 1990s, a joint United Nations committee promoting parasite vaccines shortlisted promising candidates including for schistosomiasis discussed below. After examining the complexity of immune responses in human hosts infected with schistosomes, we review and discuss the antigen design and preclinical and clinical development of the four leading vaccine candidates: Sm-TSP-2 in Phase 1b/2b, Sm14 in Phase 2a/2b, Sm-p80 in Phase 1 preparation, and Sh28GST in Phase 3. Our assessment of currently leading vaccine candidates revealed some methodological issues that preclude a fair comparison between candidates and the rationale to advance in clinical development. These include (1) variability in animal models - in particular non-human primate studies - and predictive values of each for protection in humans; (2) lack of consensus on the assessment of parasitological and immunological parameters; (3) absence of reliable surrogate markers of protection; (4) lack of well-designed parasitological and immunological natural history studies in the context of mass drug administration with praziquantel. The controlled human infection model - while promising and unique - requires validation against efficacy outcomes in endemic settings. Further research is also needed on the impact of advanced adjuvants targeting specific parts of the innate immune system that may induce potent, protective and durable immune responses with the ultimate goal of achieving meaningful worm reduction.
Collapse
Affiliation(s)
- Ursula Panzner
- International Vaccine Institute, Seoul, South Korea
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | | | | | - Florian Marks
- International Vaccine Institute, Seoul, South Korea
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- University of Antananarivo, Antananarivo, Madagascar
| | | | - Afzal A. Siddiqui
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
8
|
Qokoyi NK, Masamba P, Kappo AP. Proteins as Targets in Anti-Schistosomal Drug Discovery and Vaccine Development. Vaccines (Basel) 2021; 9:762. [PMID: 34358178 PMCID: PMC8310332 DOI: 10.3390/vaccines9070762] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 01/23/2023] Open
Abstract
Proteins hardly function in isolation; they form complexes with other proteins or molecules to mediate cell signaling and control cellular processes in various organisms. Protein interactions control mechanisms that lead to normal and/or disease states. The use of competitive small molecule inhibitors to disrupt disease-relevant protein-protein interactions (PPIs) holds great promise for the development of new drugs. Schistosome invasion of the human host involves a variety of cross-species protein interactions. The pathogen expresses specific proteins that not only facilitate the breach of physical and biochemical barriers present in skin, but also evade the immune system and digestion of human hemoglobin, allowing for survival in the host for years. However, only a small number of specific protein interactions between the host and parasite have been functionally characterized; thus, in-depth understanding of the molecular mechanisms of these interactions is a key component in the development of new treatment methods. Efforts are now focused on developing a schistosomiasis vaccine, as a proposed better strategy used either alone or in combination with Praziquantel to control and eliminate this disease. This review will highlight protein interactions in schistosomes that can be targeted by specific PPI inhibitors for the design of an alternative treatment to Praziquantel.
Collapse
Affiliation(s)
| | | | - Abidemi Paul Kappo
- Molecular Biophysics and Structural Biology (MBSB) Group, Department of Biochemistry, Kingsway Campus, University of Johannesburg, Auckland Park 2006, South Africa; (N.K.Q.); (P.M.)
| |
Collapse
|
9
|
Al-Naseri A, Al-Absi S, El Ridi R, Mahana N. A comprehensive and critical overview of schistosomiasis vaccine candidates. J Parasit Dis 2021; 45:557-580. [PMID: 33935395 PMCID: PMC8068781 DOI: 10.1007/s12639-021-01387-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/31/2021] [Indexed: 12/11/2022] Open
Abstract
A digenetic platyhelminth Schistosoma is the causative agent of schistosomiasis, one of the neglected tropical diseases that affect humans and animals in numerous countries in the Middle East, sub-Saharan Africa, South America and China. Several control methods were used for prevention of infection or treatment of acute and chronic disease. Mass drug administration led to reduction in heavy-intensity infections and morbidity, but failed to decrease schistosomiasis prevalence and eliminate transmission, indicating the need to develop anti-schistosome vaccine to prevent infection and parasite transmission. This review summarizes the efficacy and protective capacity of available schistosomiasis vaccine candidates with some insights and future prospects.
Collapse
Affiliation(s)
- Aya Al-Naseri
- Zoology Department, Faculty of Science, Cairo Univesity, Giza, 12613 Egypt
| | - Samar Al-Absi
- Zoology Department, Faculty of Science, Cairo Univesity, Giza, 12613 Egypt
| | - Rashika El Ridi
- Zoology Department, Faculty of Science, Cairo Univesity, Giza, 12613 Egypt
| | - Noha Mahana
- Zoology Department, Faculty of Science, Cairo Univesity, Giza, 12613 Egypt
| |
Collapse
|
10
|
Molehin AJ, Gray SA, Turner C, Davis J, Zhang W, Khatoon S, Rattan M, Kernen R, Peterson C, Sennoune SR, Carter D, Siddiqui AA. Process Development of Sj-p80: A Low-Cost Transmission-Blocking Veterinary Vaccine for Asiatic Schistosomiasis. Front Immunol 2021; 11:578715. [PMID: 33732227 PMCID: PMC7959798 DOI: 10.3389/fimmu.2020.578715] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 12/30/2020] [Indexed: 11/16/2022] Open
Abstract
Asiatic schistosomiasis caused by Schistosoma japonicum is a neglected tropical disease resulting in significant morbidity to both humans and animals - particularly bovines - in endemic areas. Infection with this parasite leads to less healthy herds, causing problems in communities which rely on bovines for farming, milk and meat production. Additionally, excretion of parasite eggs in feces perpetuates the life cycle and can lead to human infection. We endeavored to develop a minimally purified, inexpensive, and effective vaccine based on the 80 kDa large subunit of the calcium activated neutral protease (calpain) from S. japonicum (Sj-p80). Here we describe the production of veterinary vaccine-grade Sj-p80 at four levels of purity and demonstrate in a pilot study that minimally purified antigen provides protection against infection in mice when paired with a low-cost veterinary adjuvant, Montanide™ ISA61 VG. Preliminary data demonstrate that the vaccine is immunogenic with robust antibody titers following immunization, and vaccination resulted in a reduction of parasite eggs being deposited in the liver (23.4-51.4%) and intestines (1.9-55.1%) depending on antigen purity as well as reducing the ability of these eggs to hatch into miracidia by up to 31.6%. We therefore present Sj-p80 as a candidate vaccine antigen for Asiatic schistosomiasis which is now primed for continued development and testing in bovines in endemic areas. A successful bovine vaccine could play a major role in reducing pathogen transmission to humans by interrupting the parasitic life cycle and improving quality of life for people living in endemic countries.
Collapse
Affiliation(s)
- Adebayo J. Molehin
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Sean A. Gray
- PAI Life Sciences Inc, Seattle, WA, United States
| | - Cheri Turner
- PAI Life Sciences Inc, Seattle, WA, United States
| | | | - Weidong Zhang
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Sabiha Khatoon
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Madison Rattan
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Rebecca Kernen
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Christopher Peterson
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Souad R. Sennoune
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | | | - Afzal A. Siddiqui
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
11
|
Perera DJ, Hassan AS, Jia Y, Ricciardi A, McCluskie MJ, Weeratna RD, Ndao M. Adjuvanted Schistosoma mansoni-Cathepsin B With Sulfated Lactosyl Archaeol Archaeosomes or AddaVax™ Provides Protection in a Pre-Clinical Schistosomiasis Model. Front Immunol 2020; 11:605288. [PMID: 33304354 PMCID: PMC7701121 DOI: 10.3389/fimmu.2020.605288] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/20/2020] [Indexed: 12/15/2022] Open
Abstract
Schistosomiasis threatens 800 million people worldwide. Chronic pathology manifests as hepatosplenomegaly, and intestinal schistosomiasis caused by Schistosoma mansoni can lead to liver fibrosis, cirrhosis, and blood in the stool. To assist the only FDA-approved drug, praziquantel, in parasite elimination, the development of a vaccine would be of high value. S. mansoni Cathepsin B (SmCB) is a well-documented vaccine target for intestinal schistosomiasis. Herein, we test the increased efficacy and immunogenicity of SmCB when combined with sulfated lactosyl archaeol (SLA) archaeosomes or AddaVax™ (a squalene based oil-in-water emulsion). Both vaccine formulations resulted in robust humoral and cell mediated immune responses. Impressively, both formulations were able to reduce parasite burden greater than 40% (WHO standard), with AddaVax™ reaching 86.8%. Additionally, SmCB with both adjuvants were able to reduce granuloma size and the amount of larval parasite hatched from feces, which would reduce transmission. Our data support SmCB as a target for S. mansoni vaccination; especially when used in an adjuvanted formulation.
Collapse
Affiliation(s)
- Dilhan J Perera
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada.,Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Adam S Hassan
- Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Yimei Jia
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Alessandra Ricciardi
- Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Michael J McCluskie
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Risini D Weeratna
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Momar Ndao
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada.,Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada.,National Reference Center for Parasitology, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
12
|
Current status and future prospects of protein vaccine candidates against Schistosoma mansoni infection. Parasite Epidemiol Control 2020; 11:e00176. [PMID: 32923703 PMCID: PMC7475110 DOI: 10.1016/j.parepi.2020.e00176] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 06/04/2020] [Accepted: 08/16/2020] [Indexed: 12/30/2022] Open
Abstract
Schistosomiasis is an acute and chronic tropical parasitic disease caused by blood dwelling worm of the genus Schistosoma. It is the most destructive disease globally and is a major cause of morbidity and mortality for developing countries. Three main species of schistosomes infect human beings from which S. mansoni is the most common and widespread. Over the last several decades, chemotherapy using praziquantel has been a commonly used strategy for the treatment and control of schistosomiasis. However, control programs focused exclusively on chemotherapy have been challenging because of the frequency and rapidity of reinfection and these programs were expensive. Thus, new schistosomiasis control strategies will be needed. Vaccination strategy would be an ideal tool for a significant and sustainable reduction in the transmission and disease burden of schistosomiasis. An effective anti schistosome vaccine would greatly contribute to decreasing schistosomiasis-associated morbidity via protective immune responses leading to reduced worm burdens and decreased egg production. Vaccine development is a long process that can take decades. There have been three candidate vaccines that have been produced by Good Manufacturing Procedure and entered human clinical trials for S. mansoni are Sm14, SmTSP-2, and Sm-p80. Other candidates that are in pre-clinical trials at various stages include paramyosin, Sm29, SmKI-1, and Sm23. Since the growth of several new technologies, including genomics, transcriptomics, microarrays, immunomic profiling, and proteomics, have helped in the identification of promising new target schistosome antigens. Therefore, this review considers the present status of protein vaccine candidates against Schistosoma mansoni and provides some insight on prospects vaccine design and discovery.
Collapse
Key Words
- AE, Asparaginyl Endopeptidase
- Ab, Antibody
- Ag, Antigen
- CB, Cathepsin B
- CD, Cathepsin D
- CL3, Cathepsin L3
- DNA, Deoxyribonucleic Acid
- FA, Fatty Acid
- FABP, Fatty Acid Binding Protein
- GLA-Alum, Glucopyranosyl Lipid A Formulated in Aluminum
- GLA-SE, Glucopyranosyl Lipid Adjuvant Stable Emulsion
- IFN-γ, Interferon Gamma
- IL, Interleukin
- Ig, Immunoglobulin
- KI, Kunitz Type Protease Inhibitor
- LcP, Lipid Core Peptide
- Pmy, Paramyosin
- Protein vaccine
- Schistosoma mansoni
- Schistosomiasis
- Sm, Schistosoma mansoni
- TSP, Tetraspanins
- Th, T-helper Cells
- Vaccine candidates
- WHO, World Health Organization
Collapse
|
13
|
Wesołowska A, Basałaj K, Zawistowska-Deniziak A, Januszkiewicz K, Kozak Ljunggren M, Jedlina L, Wędrychowicz H. The failure of a DNA prime/protein boost regime and CTLA-4 mediated targeting to improve the potency of a DNA vaccine encoding Fasciola hepatica phosphoglycerate kinase in sheep. Vet Immunol Immunopathol 2019; 217:109941. [PMID: 31550657 DOI: 10.1016/j.vetimm.2019.109941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 09/06/2019] [Accepted: 09/08/2019] [Indexed: 11/15/2022]
Abstract
DNA vaccination in large animals has often been associated with poor immunogenicity, consequently several approaches have been evaluated to enhance its efficacy. Here, we tested a cDNA encoding a phosphoglycerate kinase from Fasciola hepatica (cDNA-FhPGK/pCMV) as a vaccine against ovine fasciolosis and investigated whether a DNA prime/protein boost regime or CTLA-4 (cytotoxic lymphocyte antigen 4) mediated targeting improved DNA vaccine efficacy. No statistically significant differences in the cellular responses were seen in either vaccine trial when compared with the respective control groups. However, specific antibody responses were considerably enhanced in DNA primed/protein boosted sheep, but not among CTLA-4 targeted cDNA-FhPGK/pCMV vaccinated animals. Nevertheless, increased titers of specific IgG1 did not contribute to protection against infection, with no differences in liver fluke recoveries reported. If DNA vaccines against fasciolosis in target species are to reach the market one day, more research in this area is needed.
Collapse
Affiliation(s)
- Agnieszka Wesołowska
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Twarda 51/55, 00-818 Warsaw, Poland.
| | - Katarzyna Basałaj
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Twarda 51/55, 00-818 Warsaw, Poland
| | - Anna Zawistowska-Deniziak
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Twarda 51/55, 00-818 Warsaw, Poland
| | - Kamil Januszkiewicz
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Twarda 51/55, 00-818 Warsaw, Poland
| | - Monika Kozak Ljunggren
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Twarda 51/55, 00-818 Warsaw, Poland
| | - Luiza Jedlina
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Twarda 51/55, 00-818 Warsaw, Poland
| | - Halina Wędrychowicz
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Twarda 51/55, 00-818 Warsaw, Poland
| |
Collapse
|
14
|
Zhang W, Molehin AJ, Rojo JU, Sudduth J, Ganapathy PK, Kim E, Siddiqui AJ, Freeborn J, Sennoune SR, May J, Lazarus S, Nguyen C, Redman WK, Ahmad G, Torben W, Karmakar S, Le L, Kottapalli KR, Kottapalli P, Wolf RF, Papin JF, Carey D, Gray SA, Bergthold JD, Damian RT, Mayer BT, Marks F, Reed SG, Carter D, Siddiqui AA. Sm-p80-based schistosomiasis vaccine: double-blind preclinical trial in baboons demonstrates comprehensive prophylactic and parasite transmission-blocking efficacy. Ann N Y Acad Sci 2018; 1425:38-51. [PMID: 30133707 PMCID: PMC6110104 DOI: 10.1111/nyas.13942] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/06/2018] [Accepted: 07/10/2018] [Indexed: 01/01/2023]
Abstract
Schistosomiasis is of public health importance to an estimated one billion people in 79 countries. A vaccine is urgently needed. Here, we report the results of four independent, double-blind studies of an Sm-p80-based vaccine in baboons. The vaccine exhibited potent prophylactic efficacy against transmission of Schistosoma mansoni infection and was associated with significantly less egg-induced pathology, compared with unvaccinated control animals. Specifically, the vaccine resulted in a 93.45% reduction of pathology-producing female worms and significantly resolved the major clinical manifestations of hepatic/intestinal schistosomiasis by reducing the tissue egg-load by 89.95%. A 35-fold decrease in fecal egg excretion in vaccinated animals, combined with an 81.51% reduction in hatching of eggs into the snail-infective stage (miracidia), demonstrates the parasite transmission-blocking potential of the vaccine. Substantially higher Sm-p80 expression in female worms and Sm-p80-specific antibodies in vaccinated baboons appear to play an important role in vaccine-mediated protection. Preliminary analyses of RNA sequencing revealed distinct molecular signatures of vaccine-induced effects in baboon immune effector cells. This study provides comprehensive evidence for the effectiveness of an Sm-p80-based vaccine for schistosomiasis.
Collapse
Affiliation(s)
- Weidong Zhang
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Adebayo J. Molehin
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Juan U. Rojo
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH
| | - Justin Sudduth
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Pramodh K. Ganapathy
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Eunjee Kim
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Arif J. Siddiqui
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Jasmin Freeborn
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Souad R. Sennoune
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Jordan May
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Samra Lazarus
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Catherine Nguyen
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Whitni K. Redman
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Gul Ahmad
- Department of Natural Sciences, Peru State College, Peru, NE
| | | | - Souvik Karmakar
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Loc Le
- Biomedical Research Institute, Rockville, MD
| | | | | | - Roman F. Wolf
- Oklahoma City VA Health Care System, Oklahoma City, OK
| | - James F. Papin
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - David Carey
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | | | | | - Raymond T. Damian
- Department of Cellular Biology, University of Georgia, Athens, Georgia
| | - Bryan T. Mayer
- Vaccine Immunology Statistical Center, Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Florian Marks
- International Vaccine Institute SNU Research Park, Seoul, South Korea
- The Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | - Darrick Carter
- PAI Life Sciences, Seattle, Washington, WA
- Infectious Disease Research Institute, Seattle, WA
| | - Afzal A. Siddiqui
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| |
Collapse
|
15
|
Zhang W, Ahmad G, Molehin AJ, Torben W, Le L, Kim E, Lazarus S, Siddiqui AJ, Carter D, Siddiqui AA. Schistosoma mansoni antigen Sm-p80: prophylactic efficacy using TLR4 agonist vaccine adjuvant glucopyranosyl lipid A-Alum in murine and non-human primate models. J Investig Med 2018; 66:1124-1132. [PMID: 29997146 PMCID: PMC6288690 DOI: 10.1136/jim-2018-000786] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2018] [Indexed: 01/06/2023]
Abstract
Sm-p80, the large subunit of Schistosoma mansoni calpain, is a leading candidate for a schistosomiasis vaccine. The prophylactic and antifecundity efficacy of Sm-p80 has been tested in three animal models (mouse, hamster and baboon) using a multitude of vaccine formulations and approaches. In our continual effort to enhance the vaccine efficacy, in this study, we have utilized the adjuvant, synthetic hexa-acylated lipid A derivative, glucopyranosyl lipid A (GLA) formulated in aluminum (GLA-Alum) with recombinant Sm-p80. The rSm-p80+GLA-Alum immunization regimen provided 33.33%–53.13% reduction in worm burden in the mouse model and 38% worm burden reduction in vaccinated baboons. Robust Sm-p80-specific immunoglobulin (Ig)G, IgG1, IgG2a and IgM responses were observed in all immunized animals. The rSm-p80+GLA-Alum coadministration induced a mix of T-helper (Th) cells (Th1, Th2 and Th17) responses as determined via the release of interleukin (IL)-2, IL-4, IL-18, IL-21, IL-22 and interferon-γ.
Collapse
Affiliation(s)
- Weidong Zhang
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas, USA.,Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Gul Ahmad
- Department of Natural Sciences, School of Arts & Sciences, Peru State College, Peru, Nebraska, USA
| | - Adebayo J Molehin
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas, USA.,Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Workineh Torben
- Comparative Pathology/Immunology, Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Loc Le
- Bladder Immunology Group, Biomedical Research Institute, Rockville, Maryland, USA
| | - Eunjee Kim
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas, USA.,Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Samra Lazarus
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas, USA.,Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Arif J Siddiqui
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas, USA.,Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | | | - Afzal A Siddiqui
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas, USA.,Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| |
Collapse
|
16
|
Siddiqui AJ, Molehin AJ, Zhang W, Ganapathy PK, Kim E, Rojo JU, Redman WK, Sennoune SR, Sudduth J, Freeborn J, Hunter D, Kottapalli KR, Kottapalli P, Wettashinghe R, van Dam GJ, Corstjens PLAM, Papin JF, Carey D, Torben W, Ahmad G, Siddiqui AA. Sm-p80-based vaccine trial in baboons: efficacy when mimicking natural conditions of chronic disease, praziquantel therapy, immunization, and Schistosoma mansoni re-encounter. Ann N Y Acad Sci 2018; 1425:19-37. [PMID: 29888790 DOI: 10.1111/nyas.13866] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/27/2018] [Accepted: 05/02/2018] [Indexed: 11/28/2022]
Abstract
Sm-p80-based vaccine efficacy for Schistosoma mansoni was evaluated in a baboon model of infection and disease. The study was designed to replicate a human vaccine implementation scenario for endemic regions in which vaccine would be administered following drug treatment of infected individuals. In our study, the Sm-p80-based vaccine reduced principal pathology producing hepatic egg burdens by 38.0% and egg load in small and large intestines by 72.2% and 49.4%, respectively, in baboons. Notably, hatching rates of eggs recovered from liver and small and large intestine of vaccinated animals were significantly reduced, by 60.4%, 48.6%, and 82.3%, respectively. Observed reduction in egg maturation/hatching rates was supported by immunofluorescence and confocal microscopy showing unique differences in Sm-p80 expression in worms of both sexes and matured eggs. Vaccinated baboons had a 64.5% reduction in urine schistosome circulating anodic antigen, a parameter that reflects worm numbers/health status in infected hosts. Preliminary analyses of RNA sequencing revealed unique genes and canonical pathways associated with establishment of chronic disease, praziquantel-mediated parasite killing, and Sm-p80-mediated protection in vaccinated baboons. Overall, our study demonstrated efficacy of the Sm-p80 vaccine and provides insight into some of the epistatic interactions associated with protection.
Collapse
Affiliation(s)
- Arif J Siddiqui
- School of Medicine, Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Adebayo J Molehin
- School of Medicine, Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Weidong Zhang
- School of Medicine, Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Pramodh K Ganapathy
- School of Medicine, Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Eunjee Kim
- School of Medicine, Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Juan U Rojo
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, New Hampshire
| | - Whitni K Redman
- School of Medicine, Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Souad R Sennoune
- School of Medicine, Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Justin Sudduth
- School of Medicine, Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Jasmin Freeborn
- School of Medicine, Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Derick Hunter
- School of Medicine, Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | | | - Pratibha Kottapalli
- Center for Biotechnology and Genomics, Texas Tech University, Lubbock, Texas
| | | | - Govert J van Dam
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Paul L A M Corstjens
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - James F Papin
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - David Carey
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Workineh Torben
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana
| | - Gul Ahmad
- Department of Biology, School of Arts & Sciences, Peru State College, Peru, Nebraska
| | - Afzal A Siddiqui
- School of Medicine, Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| |
Collapse
|
17
|
Schistosoma egg-induced liver pathology resolution by Sm-p80-based schistosomiasis vaccine in baboons. Pathology 2018; 50:442-449. [PMID: 29739616 DOI: 10.1016/j.pathol.2018.01.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/22/2017] [Accepted: 01/11/2018] [Indexed: 01/20/2023]
Abstract
Schistosomiasis remains a serious chronic debilitating hepato-intestinal disease. Current control measures based on mass drug administration are inadequate due to sustained re-infection rates, low treatment coverage and emergence of drug resistance. Hence, there is an urgent need for a schistosomiasis vaccine for disease control. In this study, we assessed the anti-pathology efficacy of Schistosoma mansoni large subunit of calpain (Sm-p80)-based vaccine against schistosomiasis caused by infections with Schistosoma mansoni in baboons. We also evaluated the disease transmission-blocking potential of Sm-p80 vaccine. Immunisations with Sm-p80-based vaccine resulted in significant reduction of hepatic egg load in vaccinated baboons (67.7% reduction, p = 0.0032) when compared to the control animals, indicative of reduction in pathology. There was also a significant reduction in sizes of egg-induced granulomas in baboons immunised with Sm-p80 vaccine compared to their control counterparts. Egg hatching rate analysis revealed an overall 85.6% reduction (p = 0.0018) in vaccinated animals compared to the controls, highlighting the potential role of Sm-p80 vaccine in disease transmission. The findings on anti-pathology efficacy and transmission-blocking potential presented in this study have formed the basis for a large-scale double-blinded baboon experiment that is currently underway.
Collapse
|
18
|
Stutzer C, Richards SA, Ferreira M, Baron S, Maritz-Olivier C. Metazoan Parasite Vaccines: Present Status and Future Prospects. Front Cell Infect Microbiol 2018; 8:67. [PMID: 29594064 PMCID: PMC5859119 DOI: 10.3389/fcimb.2018.00067] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/26/2018] [Indexed: 12/21/2022] Open
Abstract
Eukaryotic parasites and pathogens continue to cause some of the most detrimental and difficult to treat diseases (or disease states) in both humans and animals, while also continuously expanding into non-endemic countries. Combined with the ever growing number of reports on drug-resistance and the lack of effective treatment programs for many metazoan diseases, the impact that these organisms will have on quality of life remain a global challenge. Vaccination as an effective prophylactic treatment has been demonstrated for well over 200 years for bacterial and viral diseases. From the earliest variolation procedures to the cutting edge technologies employed today, many protective preparations have been successfully developed for use in both medical and veterinary applications. In spite of the successes of these applications in the discovery of subunit vaccines against prokaryotic pathogens, not many targets have been successfully developed into vaccines directed against metazoan parasites. With the current increase in -omics technologies and metadata for eukaryotic parasites, target discovery for vaccine development can be expedited. However, a good understanding of the host/vector/pathogen interface is needed to understand the underlying biological, biochemical and immunological components that will confer a protective response in the host animal. Therefore, systems biology is rapidly coming of age in the pursuit of effective parasite vaccines. Despite the difficulties, a number of approaches have been developed and applied to parasitic helminths and arthropods. This review will focus on key aspects of vaccine development that require attention in the battle against these metazoan parasites, as well as successes in the field of vaccine development for helminthiases and ectoparasites. Lastly, we propose future direction of applying successes in pursuit of next generation vaccines.
Collapse
Affiliation(s)
- Christian Stutzer
- Tick Vaccine Group, Department of Genetics, University of Pretoria, Pretoria, South Africa
| | | | | | | | | |
Collapse
|
19
|
Mekonnen GG, Pearson M, Loukas A, Sotillo J. Extracellular vesicles from parasitic helminths and their potential utility as vaccines. Expert Rev Vaccines 2018; 17:197-205. [PMID: 29353519 DOI: 10.1080/14760584.2018.1431125] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Helminths are multicellular parasites affecting nearly three billion people worldwide. To orchestrate a parasitic existence, helminths secrete different molecules, either in soluble form or contained within extracellular vesicles (EVs). EVs are secreted by most cell types and organisms, and have varied roles in intercellular communication, including immune modulation and pathogenesis. AREAS COVERED In this review, we describe the nucleic acid and proteomic composition of EVs from helminths, with a focus on the protein vaccine candidates present on the EV surface membrane, and discuss the potential utility of helminth EVs and their constituent proteins in the fight against helminth infections. EXPERT COMMENTARY A significant number of proteins present in helminth-secreted EVs are known vaccine candidates. The characterization of helminth EV proteomes will shed light on host-pathogen interactions, facilitate the discovery of new diagnostic biomarkers, and provide a novel approach for the development of new control measures against helminth infections.
Collapse
Affiliation(s)
- Gebeyaw Getnet Mekonnen
- a Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia.,b Department of Medical Parasitology , School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar , Gondar , Ethiopia
| | - Mark Pearson
- a Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| | - Alex Loukas
- a Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| | - Javier Sotillo
- a Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| |
Collapse
|
20
|
Ranasinghe SL, Duke M, Harvie M, McManus DP. Kunitz-type protease inhibitor as a vaccine candidate against schistosomiasis mansoni. Int J Infect Dis 2018; 66:26-32. [DOI: 10.1016/j.ijid.2017.10.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/26/2017] [Accepted: 10/30/2017] [Indexed: 01/08/2023] Open
|
21
|
Torben W, Molehin AJ, Blair RV, Kenway C, Shiro F, Roslyn D, Chala B, Gutu D, Kebede MA, Ahmad G, Zhang W, Aye P, Mohan M, Lackner A, Siddiqui AA. The self-curing phenomenon of schistosome infection in rhesus macaques: insight from in vitro studies. Ann N Y Acad Sci 2017; 1408:79-89. [PMID: 29239481 DOI: 10.1111/nyas.13565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/25/2017] [Accepted: 10/31/2017] [Indexed: 12/26/2022]
Abstract
A reduction in the burden of schistosomiasis is potentially achievable by integrating a schistosomiasis vaccine with current control measures. Here, we determine parasite-specific in vitro responses of B, T, and NK cells from naive uninfected rhesus macaques to Schistosoma mansoni (Sm) egg (SmEA) and worm antigen (SmWA) preparations isolated from infected baboons. Pronounced B cell responses to SmEA and NK cell responses to both SmEA and SmWA were observed. High levels of IL-2 and IL-21 responses against Sm antigens were observed in T and non-T cells of lymph nodes (LNs) and gut lamina propria-derived lymphocytes (LPLs). Data analysis showed multifunctionality of LN-derived CD4+ , CD8+ , and CD4+ CD8+ double positive T cells against either SmWA or SmWA+SmEA antigen preparations. Distinct SmEA-specific multifunctional responses were observed in gut LPLs, suggesting simultaneous responses against egg antigens. These data provide insight into the immune effectors involved in schistosome responses by rhesus macaques.
Collapse
Affiliation(s)
- Workineh Torben
- Tulane National Primate Research Center, TNPRC, Comparative Pathology/Immunology, Tulane University Health Sciences Center, Covington, Louisiana
| | - Adebayo J Molehin
- Center for Tropical Medicine and Infectious Diseases, Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Robert V Blair
- Tulane National Primate Research Center, TNPRC, Comparative Pathology/Immunology, Tulane University Health Sciences Center, Covington, Louisiana
| | - Carys Kenway
- Tulane National Primate Research Center, TNPRC, Comparative Pathology/Immunology, Tulane University Health Sciences Center, Covington, Louisiana
| | - Faith Shiro
- Tulane National Primate Research Center, TNPRC, Comparative Pathology/Immunology, Tulane University Health Sciences Center, Covington, Louisiana
| | - Davis Roslyn
- Tulane National Primate Research Center, TNPRC, Comparative Pathology/Immunology, Tulane University Health Sciences Center, Covington, Louisiana
| | - Bayissa Chala
- Department of Applied Biology, Adama Science and Technology University, School of Applied Natural Sciences, Adama, Ethiopia
| | - Dereje Gutu
- Department of Veterinary Medicine, Jimma University, Jimma, Ethiopia
| | - Michael A Kebede
- Department of Epidemiology & Biostatistics, George Washington University, Washington, DC
| | - Gul Ahmad
- Department of Biology, Peru State College, Peru, Nebraska
| | - Weidong Zhang
- Center for Tropical Medicine and Infectious Diseases, Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Pyone Aye
- Tulane National Primate Research Center, TNPRC, Comparative Pathology/Immunology, Tulane University Health Sciences Center, Covington, Louisiana
| | - Mahesh Mohan
- Tulane National Primate Research Center, TNPRC, Comparative Pathology/Immunology, Tulane University Health Sciences Center, Covington, Louisiana
| | - Andrew Lackner
- Tulane National Primate Research Center, TNPRC, Comparative Pathology/Immunology, Tulane University Health Sciences Center, Covington, Louisiana
| | - Afzal A Siddiqui
- Center for Tropical Medicine and Infectious Diseases, Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| |
Collapse
|
22
|
Schistosome vaccines: problems, pitfalls and prospects. Emerg Top Life Sci 2017; 1:641-650. [PMID: 33525844 DOI: 10.1042/etls20170094] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 12/14/2022]
Abstract
Human schistosomiasis caused by parasitic flatworms of the genus Schistosoma remains an important public health problem in spite of concerted efforts at control. An effective vaccine would be a useful addition to control strategies that currently rely on chemotherapy, but such a product is not imminent. In this review, likely causes for the lack of progress are first considered. These include the strategies used by worms to evade the immune response, concepts that have misdirected the field, an emphasis on internal antigens, and the use of the laboratory mouse for vaccine testing. On a positive note, recent investigations on self-cure by the rhesus macaque offer the most promising context for vaccine development. The identification of proteins at the parasite-host interface, especially those of the esophageal glands involved in blood processing, has provided an entirely new category of vaccine candidates that merit evaluation.
Collapse
|
23
|
Molehin AJ, Sennoune SR, Zhang W, Rojo JU, Siddiqui AJ, Herrera KA, Johnson L, Sudduth J, May J, Siddiqui AA. Cross-species prophylactic efficacy of Sm-p80-based vaccine and intracellular localization of Sm-p80/Sm-p80 ortholog proteins during development in Schistosoma mansoni, Schistosoma japonicum, and Schistosoma haematobium. Parasitol Res 2017; 116:3175-3188. [PMID: 29026995 PMCID: PMC5660642 DOI: 10.1007/s00436-017-5634-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 09/25/2017] [Indexed: 10/18/2022]
Abstract
Schistosomiasis remains a major global health problem. Despite large-scale schistosomiasis control efforts, clear limitations such as possible emergence of drug resistance and reinfection rates highlight the need for an effective schistosomiasis vaccine. Schistosoma mansoni large subunit of calpain (Sm-p80)-based vaccine formulations have shown remarkable efficacy in protecting against S. mansoni challenge infections in mice and baboons. In this study, we evaluated the cross-species protective efficacy of Sm-p80 vaccine against S. japonicum and S. haematobium challenge infections in rodent models. We also elucidated the expression of Sm-p80 and Sm-p80 ortholog proteins in different developmental stages of S. mansoni, S. haematobium, and S. japonicum. Immunization with Sm-p80 vaccine reduced worm burden by 46.75% against S. japonicum challenge infection in mice. DNA prime/protein boost (1 + 1 dose administered on a single day) resulted in 26.95% reduction in worm burden in S. haematobium-hamster infection/challenge model. A balanced Th1 (IFN-γ, TNF-α, IL-2, and IL-12) and Th2 (IL-4, IgG1) type of responses were observed following vaccination in both S. japonicum and S. haematobium challenge trials and these are associated with the prophylactic efficacy of Sm-p80 vaccine. Immunohistochemistry demonstrated that Sm-p80/Sm-p80 ortholog proteins are expressed in different life cycle stages of the three major human species of schistosomes studied. The data presented in this study reinforce the potential of Sm-p80-based vaccine for both hepatic/intestinal and urogenital schistosomiasis occurring in different geographical areas of the world. Differential expression of Sm-p80/Sm-p80 protein orthologs in different life cycle makes this vaccine potentially useful in targeting different levels of infection, disease, and transmission.
Collapse
Affiliation(s)
- Adebayo J Molehin
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Souad R Sennoune
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Weidong Zhang
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Juan U Rojo
- College of Life Sciences and Agriculture, University of New Hampshire, Durham, NH, USA
| | - Arif J Siddiqui
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Karlie A Herrera
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Laura Johnson
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Justin Sudduth
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Jordan May
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Afzal A Siddiqui
- Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
24
|
Floudas A, Cluxton CD, Fahel J, Khan AR, Saunders SP, Amu S, Alcami A, Fallon PG. Composition of the Schistosoma mansoni worm secretome: Identification of immune modulatory Cyclophilin A. PLoS Negl Trop Dis 2017; 11:e0006012. [PMID: 29073139 PMCID: PMC5681295 DOI: 10.1371/journal.pntd.0006012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/10/2017] [Accepted: 10/04/2017] [Indexed: 12/16/2022] Open
Abstract
The helminth Schistosoma mansoni modulates the infected host's immune system to facilitate its own survival, by producing excretory/secretory molecules that interact with a variety of the host's cell types including those of the immune system. Herein, we characterise the S. mansoni adult male worm secretome and identify 111 proteins, including 7 vaccine candidates and several molecules with potential immunomodulatory activity. Amongst the molecules present in the secretome, a 17-19kDa protein analogous to human cyclophilin A was identified. Given the ability of cyclophilin A to modulate the immune system by regulating antigen presenting cell activity, we sought to determine whether recombinant S. mansoni Cyclophilin A (rSmCypA) is capable of modulating bone-marrow derived dendritic cell (BMDC) and T cell responses under in vitro conditions. rSmCypA was enzymatically active and able to alter the pro-inflammatory cytokine profile of LPS-activated dendritic cells. rSmCypA also modulated DC function in the induction of CD4+ T cell proliferation with a preferential expansion of Treg cells. This work demonstrates the unique protein composition of the S. mansoni male worm secretome and immunomodulatory activity of S. mansoni Cyclophilin A.
Collapse
Affiliation(s)
- Achilleas Floudas
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Christopher D. Cluxton
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Julia Fahel
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Adnan R. Khan
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Sean P. Saunders
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Sylvie Amu
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Antonio Alcami
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Padraic G. Fallon
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
- * E-mail:
| |
Collapse
|
25
|
Rojo JU, Melkus MW, Kottapalli KR, Okiya OE, Sudduth J, Zhang W, Molehin AJ, Carter D, Siddiqui AA. Sm-p80-based schistosomiasis vaccine mediated epistatic interactions identified potential immune signatures for vaccine efficacy in mice and baboons. PLoS One 2017; 12:e0171677. [PMID: 28192534 PMCID: PMC5305113 DOI: 10.1371/journal.pone.0171677] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/23/2017] [Indexed: 12/22/2022] Open
Abstract
Schistosomiasis is a neglected parasitic disease of major public health concern as it affects over 250 million people in developing countries. Currently there is no licensed vaccine available against schistosomiasis. The Schistosoma mansoni calpain protein, Sm-p80, is a leading vaccine candidate now ready to move to clinical trials. In order to better assess Sm-p80 vaccine immunogenicity; here we used a systems biology approach employing RNA-sequencing to identify gene signatures and epistatic interactions following Sm-p80 vaccination in mouse and baboon models that may predict vaccine efficacy. Recombinant Sm-p80 + CpG-oligodeoxynucleotide (ODN) vaccine formulation induced both cellular and humoral immunity genes with a predominant TH1 response as well as TH2 and TH17 gene signatures. Early gene responses and gene-network interactions in mice immunized with rSm-p80 + ODN appear to be initiated through TLR4 signaling. CSF genes, S100A alarmin genes and TNFRSF genes appear to be a signature of vaccine immunogenicity/efficacy as identified by their participation in gene network interactions in both mice and baboons. These gene families may provide a basis for predicting desirable outcomes for vaccines against schistosomiasis leading to a better understanding of the immune system response to vaccination.
Collapse
Affiliation(s)
- Juan U. Rojo
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University School of Medicine, Lubbock, Texas, United States of America
| | - Michael W. Melkus
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University School of Medicine, Amarillo, Texas, United States of America
| | - Kameswara Rao Kottapalli
- Center for Biotechnology and Genomics. Texas Tech University, Lubbock, Texas, United States of America
| | - Oscar E. Okiya
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University School of Medicine, Lubbock, Texas, United States of America
| | - Justin Sudduth
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University School of Medicine, Lubbock, Texas, United States of America
| | - Weidong Zhang
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University School of Medicine, Lubbock, Texas, United States of America
- Department of Internal Medicine. Texas Tech University School of Medicine, Lubbock Texas, United States of America
| | - Adebayo J. Molehin
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University School of Medicine, Lubbock, Texas, United States of America
- Department of Internal Medicine. Texas Tech University School of Medicine, Lubbock Texas, United States of America
| | - Darrick Carter
- PAI Life Sciences, Seattle, Washington, United States of America
- Infectious Disease Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Afzal A. Siddiqui
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University School of Medicine, Lubbock, Texas, United States of America
- Department of Internal Medicine. Texas Tech University School of Medicine, Lubbock Texas, United States of America
| |
Collapse
|
26
|
Sm-p80-Based Schistosomiasis Vaccine: Preparation for Human Clinical Trials. Trends Parasitol 2016; 33:194-201. [PMID: 27865740 DOI: 10.1016/j.pt.2016.10.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 10/04/2016] [Accepted: 10/25/2016] [Indexed: 12/17/2022]
Abstract
Mass antiparasitic drug administration programs and other control strategies have made important contributions in reducing the global prevalence of helminths. Schistosomiasis, however, continues to spread to new geographic areas. The advent of a viable vaccine and its deployment, coupled with existing control efforts, is expected to make significant headway towards sustained schistosomiasis control. In 2016, Science ranked the schistosomiasis vaccine as one of the top 10 vaccines that needs to be urgently developed. A vaccine that is effective against geographically distinct forms of intestinal/hepatic and urinary disease is essential to make a meaningful impact in global reduction of the disease burden. In this opinion article, we focus on salient features of schistosomiasis vaccines in different phases of the clinical development pipeline and highlight the Sm-p80-based vaccine which is now being prepared for human clinical trials.
Collapse
|
27
|
Tebeje BM, Harvie M, You H, Loukas A, McManus DP. Schistosomiasis vaccines: where do we stand? Parasit Vectors 2016; 9:528. [PMID: 27716365 PMCID: PMC5045607 DOI: 10.1186/s13071-016-1799-4] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 09/14/2016] [Indexed: 12/20/2022] Open
Abstract
Schistosomiasis, caused mainly by S. mansoni, S. haematobium and S. japonicum, continues to be a serious tropical disease and public health problem resulting in an unacceptably high level of morbidity in countries where it is endemic. Praziquantel, the only drug currently available for treatment, is unable to kill developing schistosomes, it does not prevent re-infection and its continued extensive use may result in the future emergence of drug-resistant parasites. This scenario provides impetus for the development and deployment of anti-schistosome vaccines to be used as part of an integrated approach for the prevention, control and eventual elimination of schistosomiasis. This review considers the present status of candidate vaccines for schistosomiasis, and provides some insight on future vaccine discovery and design.
Collapse
Affiliation(s)
- Biniam Mathewos Tebeje
- QIMR Berghofer Medical Research Institute, Brisbane, Australia. .,School of Public Health, University of Queensland, Brisbane, Australia. .,Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia.
| | - Marina Harvie
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Hong You
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | | |
Collapse
|
28
|
Abstract
Schistosomiasis is a parasitic disease caused by helminths belonging to the Schistosoma genus. Approximately 700 million people are at risk of infection and 200 million people are currently infected. Schistosomiasis is the most important helminth infection, and treatment relies solely on the drug praziquantel. Worries of praziquantel resistance as well as high disease burden are only some of the justifications which support the development of a vaccine against schistosomiasis. To date, only 2 schistosome vaccines have made it into clinical trials: Sh28GST (Bilhvax) and Sm14. However, there are several vaccine candidates, such as TSP-2, sm-p8, and Sm-Cathepsin B, which are generating promising results in pre-clinical studies. Schistosomiasis vaccine development has been an uphill battle, and there are still several hurdles to overcome in the future. Fortunately, the research groups involved in the research for vaccine development have not abandoned their work. Furthermore, in the last few years, schistosomiasis has garnered some additional attention on a global scale due to its significant impact on public health.
Collapse
Affiliation(s)
- Alessandra Ricciardi
- a National Reference Center for Parasitoloy; Research Institute of the McGill University Health Center ; Montreal , Quebec , Canada.,b Department of Microbiology & Immunology ; McGill University ; Montreal , Quebec , Canada
| | - Momar Ndao
- a National Reference Center for Parasitoloy; Research Institute of the McGill University Health Center ; Montreal , Quebec , Canada.,b Department of Microbiology & Immunology ; McGill University ; Montreal , Quebec , Canada
| |
Collapse
|
29
|
Molehin AJ, Rojo JU, Siddiqui SZ, Gray SA, Carter D, Siddiqui AA. Development of a schistosomiasis vaccine. Expert Rev Vaccines 2016; 15:619-27. [PMID: 26651503 PMCID: PMC5070536 DOI: 10.1586/14760584.2016.1131127] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Schistosomiasis is a neglected tropical disease (NTD) of public health importance. Despite decades of implementation of mass praziquantel therapy programs and other control measures, schistosomiasis has not been contained and continues to spread to new geographic areas. A schistosomiasis vaccine could play an important role as part of a multifaceted control approach. With regards to vaccine development, many biological bottlenecks still exist: the lack of reliable surrogates of protection in humans; immune interactions in co-infections with other diseases in endemic areas; the potential risk of IgE responses to antigens in endemic populations; and paucity of appropriate vaccine efficacy studies in nonhuman primate models. Research is also needed on the role of modern adjuvants targeting specific parts of the innate immune system to tailor a potent and protective immune response for lead schistosome vaccine candidates with the long-term aim to achieve curative worm reduction. This review summarizes the current status of schistosomiasis vaccine development.
Collapse
Affiliation(s)
- Adebayo J. Molehin
- Department of Internal Medicine, Texas Tech University School of Medicine, Lubbock, Texas, USA
- Center of Tropical Medicine and Infectious Diseases, Texas Tech University School of Medicine, Lubbock, Texas, USA
| | - Juan U. Rojo
- Department of Internal Medicine, Texas Tech University School of Medicine, Lubbock, Texas, USA
- Center of Tropical Medicine and Infectious Diseases, Texas Tech University School of Medicine, Lubbock, Texas, USA
| | - Sabrina Z. Siddiqui
- Department of Internal Medicine, Texas Tech University School of Medicine, Lubbock, Texas, USA
- Center of Tropical Medicine and Infectious Diseases, Texas Tech University School of Medicine, Lubbock, Texas, USA
| | | | - Darrick Carter
- PAI Life Sciences, Washington, USA
- Infectious Disease Research Institute, Seattle, Washington, USA
| | - Afzal A. Siddiqui
- Department of Internal Medicine, Texas Tech University School of Medicine, Lubbock, Texas, USA
- Center of Tropical Medicine and Infectious Diseases, Texas Tech University School of Medicine, Lubbock, Texas, USA
| |
Collapse
|
30
|
Ricciardi A, Visitsunthorn K, Dalton JP, Ndao M. A vaccine consisting of Schistosoma mansoni cathepsin B formulated in Montanide ISA 720 VG induces high level protection against murine schistosomiasis. BMC Infect Dis 2016; 16:112. [PMID: 26945988 PMCID: PMC4779570 DOI: 10.1186/s12879-016-1444-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 02/24/2016] [Indexed: 01/13/2023] Open
Abstract
Background Schistosomiasis is the most important human helminth infection due to its impact on public health. The clinical manifestations are chronic and significantly decrease an individual’s quality of life. Infected individuals suffer from long-term organ pathologies including fibrosis which eventually leads to organ failure. The development of a vaccine against this parasitic disease would contribute to a long-lasting decrease in disease spectrum and transmission. Method Our group has chosen Schistosoma mansoni (Sm) cathepsin B, a peptidase involved in parasite feeding, as a prospective vaccine candidate. Our experimental formulation consisted of recombinant Sm-cathepsin B formulated in Montanide ISA 720 VG, a squalene based adjuvant containing a mannide mono-oleate emulsifier. Parasitological burden was assessed by determining adult worm, hepatic egg, and intestinal egg numbers in each mouse. Serum was used in ELISAs to evaluate production of antigen-specific antibodies, and isolated splenocytes were stimulated with the antigen for the analysis of cytokine secretion levels. Results The Sm-cathepsin B and Montanide formulation conferred protection against a challenge infection by significantly reducing all forms of parasitological burdens. Worm burden, hepatic egg burden and intestinal egg burden were decreased by 60 %, 62 %, and 56 %, respectively in immunized animals compared to controls (P = 0.0002, P < 0.0001, P = 0.0009, respectively). Immunizations with the vaccine elicited robust production of Sm-cathepsin B specific antibodies (endpoint titers = 122,880). Both antigen-specific IgG1 and IgG2c titers were observed, with the former having more elevated titers. Furthermore, splenocytes isolated from the immunized animals, compared to control animals, secreted higher levels of key Th1 cytokines, IFN-γ, IL-12, and TNF-α, as well as the Th2 cytokines IL-5 and IL-4 when stimulated with recombinant Sm-cathepsin B. The Th17 cytokine IL-17, the chemokine CCL5, and the growth factor GM-CSF were also significantly increased in the immunized animals compared to the controls. Conclusion The formulation tested in this study was able to significantly reduce all forms of parasite burden, stimulate robust production of antigen-specific antibodies, and induce a mixed Th1/Th2 response. These results highlight the potential of Sm-cathepsin B/Montanide ISA 720 VG as a vaccine candidate against schistosomiasis. Electronic supplementary material The online version of this article (doi:10.1186/s12879-016-1444-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alessandra Ricciardi
- Department of Microbiology & Immunology, McGill University, Montreal, QC, Canada.,National Reference Center for Parasitology, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | | | - John P Dalton
- Institute of Parasitology, McGill University, Montreal, QC, Canada.,School of Biological Sciences, Medical Biology Centre (MBC) Queen's University Belfast, Belfast, Northern Ireland
| | - Momar Ndao
- Department of Microbiology & Immunology, McGill University, Montreal, QC, Canada. .,National Reference Center for Parasitology, Research Institute of the McGill University Health Center, Montreal, QC, Canada.
| |
Collapse
|
31
|
Wedrychowicz H. Antiparasitic DNA vaccines in 21st century. Acta Parasitol 2015; 60:179-89. [PMID: 26203983 PMCID: PMC7088677 DOI: 10.1515/ap-2015-0026] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Revised: 12/15/2014] [Accepted: 12/17/2014] [Indexed: 11/25/2022]
Abstract
Demands for effective vaccines to control parasitic diseases of humans and livestock have been recently exacerbated by the development of resistance of most pathogenic parasites to anti-parasitic drugs. Novel genomic and proteomic technologies have provided opportunities for the discovery and improvement of DNA vaccines which are relatively easy as well as cheap to fabricate and stable at room temperatures. However, their main limitation is rather poor immunogenicity, which makes it necessary to couple the antigens with adjuvant molecules. This paper review recent advances in the development of DNA vaccines to some pathogenic protozoa and helminths. Numerous studies were conducted over the past 14 years of 21st century, employing various administration techniques, adjuvants and new immunogenic antigens to increase efficacy of DNA vaccines. Unfortunately, the results have not been rewarding. Further research is necessary using more extensive combinations of antigens; alternate delivery systems and more efficient adjuvants based on knowledge of the immunomodulatory capacities of parasitic protozoa and helminths.
Collapse
MESH Headings
- Animals
- Disease Transmission, Infectious/prevention & control
- Drug Discovery/trends
- Helminthiasis/immunology
- Helminthiasis/prevention & control
- Helminthiasis/transmission
- Helminthiasis, Animal/immunology
- Helminthiasis, Animal/prevention & control
- Helminthiasis, Animal/transmission
- Humans
- Protozoan Infections/immunology
- Protozoan Infections/prevention & control
- Protozoan Infections/transmission
- Protozoan Infections, Animal/immunology
- Protozoan Infections, Animal/prevention & control
- Protozoan Infections, Animal/transmission
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
- Vaccines, DNA/isolation & purification
Collapse
Affiliation(s)
- Halina Wedrychowicz
- Department of Molecular Biology, Laboratory of Molecular Parasitology, W. Stefański Institute Parasitology, Polish Academy of Sciences, 51/55 Twarda St., 00-818 Warsaw, Poland
| |
Collapse
|
32
|
Immuno-evasive tactics by schistosomes identify an effective allergy preventative. Exp Parasitol 2015; 153:139-50. [PMID: 25819297 DOI: 10.1016/j.exppara.2015.03.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 03/15/2015] [Accepted: 03/20/2015] [Indexed: 12/11/2022]
Abstract
Many chronic inflammatory diseases can be improved by helminth infection, but the mechanisms are poorly understood. Allergy and helminthiasis are both associated with Th2-like immune responses; thus, defining how infection with parasites leads to reduced allergy has been particularly challenging. We sought to better understand this conundrum by evaluating host-parasite interactions involved in Th2 immunity in human schistosomiasis. Immune cells were cultured with schistosomes and the effect on CD23, an IgE receptor associated with resistance in schistosomiasis, was evaluated. Cells treated with schistosomes demonstrated reduced surface CD23 levels with a parallel accumulation of soluble (s) CD23 suggesting this IgE receptor is proteolytically cleaved by the parasite. Consistent with this hypothesis, a schistosome-generated (SG)-sCD23 fragment of 15 kDa was identified. SG-sCD23 inhibited IgE from binding to CD23 and FcεRI, but lacked the ability to bind CD21. These results suggested that schistosomes target IgE-mediated immunity in immuno-evasive tactics. Based on its characteristics, we predicted that SG-sCD23 would function as an efficacious allergy preventative. Treatment of human FcεRI-transgenic mice with recombinant (r) SG-sCD23 reduced the ability of human IgE to induce an acute allergic response in vivo. In addition, an optimized form of rSG-sCD23 with an introduced point mutation at Asp258 (D258E)to stabilize IgE binding had increased efficacy compared to native rSG-sCD23. Schistosome infection may thus inhibit allergic-like protective immune responses by increasing soluble IgE decoy receptors. Allergy treatments based on this naturally occurring phenomenon may be highly effective and have fewer side effects with long-term use.
Collapse
|
33
|
Stephenson R, You H, McManus DP, Toth I. Schistosome Vaccine Adjuvants in Preclinical and Clinical Research. Vaccines (Basel) 2014; 2:654-85. [PMID: 26344751 PMCID: PMC4494218 DOI: 10.3390/vaccines2030654] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 08/08/2014] [Accepted: 08/15/2014] [Indexed: 12/16/2022] Open
Abstract
There is currently no vaccine available for human use for any parasitic infections, including the helminth disease, schistosomiasis. Despite many researchers working towards this goal, one of the focuses has been on identifying new antigenic targets. The bar to achieve protective efficacy in humans was set at a consistent induction of 40% protection or better by the World Health Organisation (WHO), and although this is a modest goal, it is yet to be reached with the six most promising schistosomiasis vaccine candidates (Sm28GST, IrV5, Sm14, paramyosin, TPI, and Sm23). Adjuvant selection has a large impact on the effectiveness of the vaccine, and the use of adjuvants to aid in the stimulation of the immune system is a critical step and a major variable affecting vaccine development. In addition to a comprehensive understanding of the immune system, level of protection and the desired immune response required, there is also a need for a standardised and effective adjuvant formulation. This review summarises the status of adjuvants that have been or are being employed in schistosomiasis vaccine development focusing on immunisation outcomes at preclinical and clinical stages.
Collapse
Affiliation(s)
- Rachel Stephenson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia.
| | - Hong You
- Molecular Parasitology Laboratory, Infectious Diseases Division, QIMR Berghofer Medical Research Institute, Brisbane, Queensland Q4006, Australia.
| | - Donald P McManus
- Molecular Parasitology Laboratory, Infectious Diseases Division, QIMR Berghofer Medical Research Institute, Brisbane, Queensland Q4006, Australia.
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia.
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
34
|
Shen J, Xu L, Liu Z, Li N, Wang L, Lv Z, Fung M, Wu Z, Sun X. Gene expression profile of LPS-stimulated dendritic cells induced by a recombinant Sj16 (rSj16) derived from Schistosoma japonicum. Parasitol Res 2014; 113:3073-83. [DOI: 10.1007/s00436-014-3973-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 05/25/2014] [Indexed: 11/28/2022]
|
35
|
Espíndola MS, Frantz FG, Soares LS, Masson AP, Tefé-Silva C, Bitencourt CS, Oliveira SC, Rodrigues V, Ramos SG, Silva CL, Faccioli LH. Combined immunization using DNA-Sm14 and DNA-Hsp65 increases CD8+ memory T cells, reduces chronic pathology and decreases egg viability during Schistosoma mansoni infection. BMC Infect Dis 2014; 14:263. [PMID: 24886395 PMCID: PMC4031977 DOI: 10.1186/1471-2334-14-263] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 05/07/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Schistosomiasis is one of the most important neglected diseases found in developing countries and affects 249 million people worldwide. The development of an efficient vaccination strategy is essential for the control of this disease. Previous work showed partial protection induced by DNA-Sm14 against Schistosoma mansoni infection, whereas DNA-Hsp65 showed immunostimulatory properties against infectious diseases, autoimmune diseases, cancer and antifibrotic properties in an egg-induced granuloma model. METHODS C57BL/6 mice received 4 doses of DNA-Sm14 (100 μg/dose) and DNA-Hsp65 (100 μg/dose), simultaneously administrated, or DNA-Sm14 alone, once a week, during four weeks. Three groups were included: 1- Control (no immunization); 2- DNA-Sm14; 3- DNA-Sm14/DNA-Hsp65. Two weeks following last immunization, animals were challenged subcutaneously with 30 cercariae. Fifteen, 48 and 69 days after infection splenocytes were collected to evaluate the number of CD8+ memory T cells (CD44(high)CD62(low)) using flow cytometry. Forty-eight days after challenge adult worms were collected by portal veins perfusion and intestines were collected to analyze the intestinal egg viability. Histological, immunohistochemical and soluble quantification of collagen and α-SMA accumulation were performed on the liver. RESULTS In the current work, we tested a new vaccination strategy using DNA-Sm14 with DNA-Hsp65 to potentiate the protection against schistosomiasis. Combined vaccination increased the number of CD8+ memory T cells and decreased egg viability on the intestinal wall of infected mice. In addition, simultaneous vaccination with DNA-Sm14/DNA-Hsp65 reduced collagen and α-SMA accumulation during the chronic phase of granuloma formation. CONCLUSION Simultaneous vaccination with DNA-Sm14/DNA-Hsp65 showed an immunostimulatory potential and antifibrotic property that is associated with the reduction of tissue damage on Schistosoma mansoni experimental infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Lúcia Helena Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av, do Café s/n 14040-903 Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
36
|
Zhang W, Ahmad G, Le L, Rojo JU, Karmakar S, Tillery KA, Torben W, Damian RT, Wolf RF, White GL, Carey DW, Carter D, Reed SG, Siddiqui AA. Longevity of Sm-p80-specific antibody responses following vaccination with Sm-p80 vaccine in mice and baboons and transplacental transfer of Sm-p80-specific antibodies in a baboon. Parasitol Res 2014; 113:2239-50. [PMID: 24728521 DOI: 10.1007/s00436-014-3879-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 03/24/2014] [Indexed: 12/20/2022]
Abstract
Based on data obtained using vaccine efficacy studies in mice, hamsters, and baboons, the credentials of Sm-p80 as a first tier vaccine candidate for schistosomiasis have been well established. Sm-p80-based vaccine formulation(s) have consistently exhibited potent prophylactic efficacy in reducing adult worm burden following cercarial challenge and induce killing of established adult worms in chronic infection. This vaccine is protective against both intestinal and urinary schistosomiasis. In this study, the longevity of Sm-p80-specific antibody responses was studied in mice and in baboons. Robust antibody titers were detected in mice for up to 60 weeks following vaccination with Sm-p80 recombinant vaccine (Sm-p80 + GLA-SE). In the follow-up experiments to our published studies, Sm-p80-specific IgG was also detected in baboons 5-8 years following the initial vaccination with an Sm-p80 DNA vaccine. In one baboon, transfer of Sm-p80-specific antibody was detected in umbilical cord blood and in the baby. These long-lasting humoral immune response data coupled with the vaccine efficacy data in rodents and nonhuman primates further strengthens the case for Sm-p80 to be moved forward through development leading to human clinical trials.
Collapse
Affiliation(s)
- Weidong Zhang
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, 3601 4th Street, Mail Stop 6591, Lubbock, TX, 79430, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Simultaneous priming with DNA encoding Sm-p80 and boosting with Sm-p80 protein confers protection against challenge infection with Schistosoma mansoni in mice. Parasitol Res 2014; 113:1195-200. [PMID: 24452916 DOI: 10.1007/s00436-014-3757-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 01/09/2014] [Indexed: 12/25/2022]
Abstract
Prophylactic efficacy of Sm-p80 was tested in the mouse model using DNA priming and boosting with protein approach. However, the novelty of the approach utilized in this study is that both the DNA priming and protein boosting was performed on a single day and no further vaccine inoculations were given to mice; the animals were challenged 1 month after the initial vaccine administration. Using this approach, significant reduction in worm burden (33 to 57 %) and marked decrease in egg retention in tissues (34 to 66%) was observed. Robust antibody titers and upregulation of cytokines (IL-1α/β, IL-12α, and IFN-γ) appears to correlate with the protection. This approach of administering vaccine on a single day could be greatly helpful in the field setting because it will eliminate the compliance issues that may arise with multiple boosters that may be required for optimal efficacy for some vaccines.
Collapse
|
38
|
Karmakar S, Zhang W, Ahmad G, Torben W, Alam MU, Le L, Damian RT, Wolf RF, White GL, Carey DW, Carter D, Reed SG, Siddiqui AA. Use of an Sm-p80-based therapeutic vaccine to kill established adult schistosome parasites in chronically infected baboons. J Infect Dis 2014; 209:1929-40. [PMID: 24436452 DOI: 10.1093/infdis/jiu031] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
No vaccines are available for human use for any parasitic infections, including the helminthic disease schistosomiasis. Sm-p80, the large subunit of Schistosoma mansoni calpain, is a leading antigen candidate for a schistosomiasis vaccine. Prophylactic and antifecundity efficacies of Sm-p80 have been tested using a variety of vaccine approaches in both rodent and nonhuman primate models. However, the therapeutic efficacy of a Sm-p80-based vaccine had not been determined. In this study, we evaluated the therapeutic efficacy of Sm-p80 by using 2 different strategies and 3 Sm-p80-based vaccine formulations in baboons. Vaccine formulations were able to decrease established adult worms by 10%-36%, reduce retention of eggs in tissues by 10%-57%, and decrease egg excretion in feces by 13%-33%, compared with control formulations. Marked differences were observed in B and T cell immune correlates between vaccinated and control animals. This is the first report of killing of established adult schistosome worms by a vaccine. In addition to distinct prophylactic efficacy of Sm-p80, this study adds to the evidence that Sm-p80 is a potentially important antigen with both substantial prophylactic and therapeutic efficacies. These data reinforce that Sm-p80 should be moved forward along the path toward human clinical trials.
Collapse
Affiliation(s)
- Souvik Karmakar
- Center for Tropical Medicine and Infectious Diseases Department of Immunology and Molecular Microbiology
| | - Weidong Zhang
- Center for Tropical Medicine and Infectious Diseases Department of Immunology and Molecular Microbiology
| | - Gul Ahmad
- Department of Natural Sciences, School of Arts and Sciences, Peru State College, Nebraska
| | - Workineh Torben
- Tulane National Primate Research Center, Covington, Louisiana
| | - Mayeen U Alam
- Center for Tropical Medicine and Infectious Diseases Department of Immunology and Molecular Microbiology
| | - Loc Le
- Center for Tropical Medicine and Infectious Diseases Department of Immunology and Molecular Microbiology
| | | | - Roman F Wolf
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Gary L White
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City
| | - David W Carey
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Darrick Carter
- Infectious Disease Research Institute PAI Life Sciences, Seattle, Washington
| | | | - Afzal A Siddiqui
- Center for Tropical Medicine and Infectious Diseases Department of Immunology and Molecular Microbiology Department of Internal Medicine Department of Pathology, Texas Tech University Health Sciences Center, Lubbock
| |
Collapse
|
39
|
Karmakar S, Zhang W, Ahmad G, Torben W, Alam MU, Le L, Damian RT, Wolf RF, White GL, Carey DW, Carter D, Reed SG, Siddiqui AA. Cross-species protection: Schistosoma mansoni Sm-p80 vaccine confers protection against Schistosoma haematobium in hamsters and baboons. Vaccine 2014; 32:1296-303. [PMID: 24397898 DOI: 10.1016/j.vaccine.2013.12.057] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 12/09/2013] [Accepted: 12/18/2013] [Indexed: 01/02/2023]
Abstract
The ability of the Schistosoma mansoni antigen, Sm-p80, to provide cross-species protection against Schistosoma haematobium challenge was evaluated in hamster and baboon models. Pronounced reduction in worm burden (48%) and in tissue egg load (64%) was observed in hamsters vaccinated with recombinant Sm-p80 admixed with glucopyranosyl lipid adjuvant-stable emulsion (GLA-SE). Similarly, in baboons, the Sm-p80/GLA-SE vaccine produced a 25% reduction in S. haematobium adult worms and decreased the egg load in the urinary bladder by 64%. A 40% and 53% reduction in fecal and urine egg output, respectively, was observed in vaccinated baboons. A balanced pro-inflammatory (Th17 and Th1) and Th2 type of response was generated after vaccination and appears indicative of augmented prophylactic efficacy. These data on cross-species protection coupled with the prophylactic, therapeutic and antifecundity efficacy against the homologous parasite, S. mansoni, reinforces Sm-p80 as a promising vaccine candidate. It is currently being prepared for GMP-compliant manufacture and for further pre-clinical development leading to human clinical trials. These results solidify the expectation that the Sm-p80 vaccine will provide relief for both the intestinal and the urinary schistosomiasis and thus will be greatly beneficial in reducing the overall burden of schistosomiasis.
Collapse
Affiliation(s)
- Souvik Karmakar
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Weidong Zhang
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Gul Ahmad
- Department of Natural Sciences, School of Arts & Sciences, Peru State College, Peru, NE 68321, USA
| | - Workineh Torben
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Mayeen U Alam
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Loc Le
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Raymond T Damian
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Roman F Wolf
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Gary L White
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - David W Carey
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Darrick Carter
- Infectious Disease Research Institute, Seattle, WA 98102, USA; PAI Life Sciences, Seattle, WA 98102, USA
| | - Steven G Reed
- Infectious Disease Research Institute, Seattle, WA 98102, USA
| | - Afzal A Siddiqui
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
40
|
Karmakar S, Zhang W, Ahmad G, Alam MU, Winn R, Torben W, Le L, Tillery KA, Siddiqui AA. Complement plays a minimal role in Sm-p80-mediated protection against Schistosoma mansoni. Hum Vaccin Immunother 2013; 10:640-7. [PMID: 24374377 DOI: 10.4161/hv.27576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Sm-p80, the large subunit of Schistosoma masoni calpain, is a leading antigen candidate for a schistosome vaccine. Prophylactic and antifecundity efficacy of Sm-p80 has been tested using a variety of vaccine approaches. However, the mechanism of Sm-p80-mediated killing is still unknown. In this study, potential role of complement in Sm-p80-mediated protection was studied using both in vitro (cobra venom factor inhibition) and in vivo using mice deficient in C3 (C3 -/-; B6.129S4-C3tm1Crr/J). In the absence of C3, Sm-p80-based vaccine was able to provide significant reduction in adult worm burden following challenge with schistosome cercariae in mice suggesting the effector functions of complement may be limited in this vaccine-induced protection.
Collapse
Affiliation(s)
- Souvik Karmakar
- Center for Tropical Medicine and Infectious Diseases; Texas Tech University Health Sciences Center; Lubbock, TX USA; Department of Immunology and Molecular Microbiology; Texas Tech University Health Sciences Center; Lubbock, TX USA
| | - Weidong Zhang
- Center for Tropical Medicine and Infectious Diseases; Texas Tech University Health Sciences Center; Lubbock, TX USA; Department of Immunology and Molecular Microbiology; Texas Tech University Health Sciences Center; Lubbock, TX USA
| | - Gul Ahmad
- Department of Natural Sciences; School of Arts & Sciences; Peru State College; Peru, NE USA
| | - Mayeen U Alam
- Center for Tropical Medicine and Infectious Diseases; Texas Tech University Health Sciences Center; Lubbock, TX USA; Department of Immunology and Molecular Microbiology; Texas Tech University Health Sciences Center; Lubbock, TX USA
| | - Richard Winn
- Center for Tropical Medicine and Infectious Diseases; Texas Tech University Health Sciences Center; Lubbock, TX USA; Department of Immunology and Molecular Microbiology; Texas Tech University Health Sciences Center; Lubbock, TX USA
| | | | - Loc Le
- Center for Tropical Medicine and Infectious Diseases; Texas Tech University Health Sciences Center; Lubbock, TX USA; Department of Immunology and Molecular Microbiology; Texas Tech University Health Sciences Center; Lubbock, TX USA
| | - Kory A Tillery
- Center for Tropical Medicine and Infectious Diseases; Texas Tech University Health Sciences Center; Lubbock, TX USA; Department of Immunology and Molecular Microbiology; Texas Tech University Health Sciences Center; Lubbock, TX USA
| | - Afzal A Siddiqui
- Center for Tropical Medicine and Infectious Diseases; Texas Tech University Health Sciences Center; Lubbock, TX USA; Department of Immunology and Molecular Microbiology; Texas Tech University Health Sciences Center; Lubbock, TX USA; Department of Pathology; Texas Tech University Health Sciences Center; Lubbock, TX USA
| |
Collapse
|
41
|
Olveda DU, Li Y, Olveda RM, Lam AK, Chau TNP, Harn DA, Williams GM, Gray DJ, Ross AGP. Bilharzia: Pathology, Diagnosis, Management and Control. TROPICAL MEDICINE & SURGERY 2013; 1:135. [PMID: 25346933 PMCID: PMC4208666 DOI: 10.4172/2329-9088.1000135] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
More than one billion people travel internationally each year and approximately 100 million to the tropics. Schistosomiasis is a neglected tropical disease caused by trematode blood flukes of the genus Schistosoma. It currently infects over 250 million people worldwide and results in approximately 25 million disability adjusted life years lost. Clinical manifestations depend on the affected organ. Subtle morbidities have also been documented including: growth retardation, anaemia and poor cognitive function in children. While schistosomiasis has been eradicated from Japan and significantly reduced in parts of China and Egypt, transmission in many other regions remains ongoing due to the wide-spread distribution of the intermediate snail host, poor sanitation, lack of health education and decreasing compliance to mass drug administration. Integrated control has significantly reduced the burden of disease in China but considerable financial capital is needed if similar results are to be duplicated elsewhere. Human vaccination is in various stages of development, and once found, will become an integral part of future control. This comprehensive review examines the epidemiology, pathology, diagnosis, clinical management, prevention and control of the disease.
Collapse
Affiliation(s)
- David U Olveda
- Griffith Health Institute, Griffith University, Gold Coast Campus, Australia
| | - Yuesheng Li
- Hunan Institute of Parasitic Diseases, WHO Collaborating Centre for Research and Control of Schistosomiasis on Lake Region, China and Queensland Institute of Medical Research, Australia
| | - Remigio M Olveda
- Research Institute for Tropical Medicine, Department of Health, Philippines
| | - Alfred K Lam
- Griffith Health Institute, Griffith University, Gold Coast Campus, Australia
| | - Thao N P Chau
- Flinders University, Discipline of Public Health, Australia
| | - Donald A Harn
- College of Veterinary Medicine, University of Georgia, USA
| | - Gail M Williams
- School of Population Health, University of Queensland, Brisbane, Australia
| | - Darren J Gray
- School of Population Health, University of Queensland, Brisbane, Australia
| | - Allen G P Ross
- Griffith Health Institute, Griffith University, Gold Coast Campus, Australia
| |
Collapse
|
42
|
Torben W, Ahmad G, Zhang W, Nash S, Le L, Karmakar S, Siddiqui AA. Role of antibody dependent cell mediated cytotoxicity (ADCC) in Sm-p80-mediated protection against Schistosoma mansoni. Vaccine 2012; 30:6753-8. [PMID: 23000221 DOI: 10.1016/j.vaccine.2012.09.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 08/07/2012] [Accepted: 09/10/2012] [Indexed: 10/27/2022]
Abstract
Schistosomiasis is a major health problem in the developing world and for international travelers to the endemic countries. Existing strategies to control schistosomiasis have had limited successes so far. The addition of an effective vaccine in existing control measures would be greatly beneficial in reducing the impact of the disease. In this regard, Sm-p80 mediated protection against intestinal schistosomiasis caused by Schistosoma mansoni has been observed to be promising in two animal models of infection and disease. In this study, the role of antibody dependent cell mediated cytotoxicity (ADCC) was deciphered in Sm-p80-mediated protection especially in the elimination of lung stage schistosomula. This was achieved using lung lavage cells and lung cells that were isolated from mice immunized with and without Sm-p80 formulated in a recombinant vaccine formulation. Significant differences were observed in cytotoxicity assays using immune sera with the lung lavage cells which showed 51% more killing of schistosomula and elevated levels of nitric oxide in the supernatants were detected compared to controls.
Collapse
Affiliation(s)
- Workineh Torben
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Liu HF, Li W, Lu MB, Yu LJ. Pharmacokinetics and risk evaluation of DNA vaccine against Schistosoma japonicum. Parasitol Res 2012; 112:59-67. [PMID: 22990210 DOI: 10.1007/s00436-012-3104-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 08/24/2012] [Indexed: 02/03/2023]
Abstract
DNA plasmid immunization is a novel approach of preventive and therapeutic vaccine. More than 100 DNA vaccines have been on preclinical or clinical phase trials, and four kinds of DNA vaccines for livestock have been approved by USDA, CFIA, and APVMA. Schistosomiasis is a worldwide parasitic disease, and vaccine immunization is supposed to be a promising approach to control the health crisis. On the basis of former preclinical studies, we further focused on the pharmacokinetics and risk evaluation of DNA vaccine in vivo. In the present study, enhanced green fluorescent protein (EGFP) report gene was fused with Schistosoma japonicum 23 kDa transmembrane protein antigen gene (Sj23) and constructed into DNA vaccine pVIVO2-Sj23.EGFP. After intramuscularly injecting 100 μg of purified DNA vaccine plasmid to immunizate BALB/c mice, we studied the tissue distribution of DNA plasmid and expressed Sj23.EGFP antigen, the persistence time of elicited antibodies, and the risk of DNA vaccine transferred into intestinal microorganisms. The results showed that DNA vaccine plasmid could be distributed into all tissues of the body after injection; however, only few organs including the injected muscle were detected DNA vaccine at postimmunization until the 100 days by PCR technology; the detection of green fluorescence protein displayed that DNA vaccine could be expressed in almost every tissue and organs; the ELISA assay indicated the immune antibody against Sj23 could persist over 70 days; and the DNA vaccine transferring intestinal flora results was negative. The results indicated that the DNA vaccine has systemic protection and long-lasting effectivity and is safe to intestinal flora.
Collapse
Affiliation(s)
- Hai-Feng Liu
- College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China.
| | | | | | | |
Collapse
|
44
|
Abstract
Schistosomiasis is a major neglected tropical disease of public health importance to a billion people. An estimated 200 million people are currently infected; an additional 779 million individuals are at risk to acquire the infection in 74 countries. Despite many years of implementation of mass anti-parasitic drug therapy programs and other control measures, this disease has not been contained and continues to spread to new geographic areas. The discovery of a protective vaccine still remains the most potentially effective means for the control of this disease, especially if the vaccine provides long-term immunity against the infection. A vaccine would contribute to the reduction of schistosomiasis morbidity through induced immune responses leading to decrease in parasite load and reduced egg production. This vaccine could be administered to children between the ages of 3 and 12 years to prevent severe infection in a particularly high risk population. This review summarizes the current status of schistosomiasis vaccine development.
Collapse
Affiliation(s)
- Afzal A Siddiqui
- Department of Microbiology and Immunology, Internal Medicine, Pathology, Texas Tech University Health Sciences Center, Lubbock, TX USA.
| | | | | |
Collapse
|
45
|
Ahmad G, Zhang W, Torben W, Ahrorov A, Damian RT, Wolf RF, White GL, Carey DW, Mwinzi PNM, Ganley-Leal L, Kennedy RC, Siddiqui AA. Preclinical prophylactic efficacy testing of Sm-p80-based vaccine in a nonhuman primate model of Schistosoma mansoni infection and immunoglobulin G and E responses to Sm-p80 in human serum samples from an area where schistosomiasis is endemic. J Infect Dis 2011; 204:1437-49. [PMID: 21921206 DOI: 10.1093/infdis/jir545] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The prophylactic efficacy of a schistosome antigen (Sm-p80) was tested in a nonhuman primate model, the baboon. Using a total of 28 baboons, different vaccination strategies were used including recombinant Sm-p80 protein formulated in Toll-like receptor 7 and Toll-like receptor 9 agonists, and DNA priming followed by boosting with protein plus adjuvants. Recombinant protein approaches provided levels of prophylactic efficacy of 52%-58%, whereas prime-boost approaches conferred 38%-47% protection in baboons. An appropriately balanced pro-inflammatory (T-helper 17 [Th17] and Th1) and anti-inflammatory (Th2) type of response was generated; the Th1 and Th17 types of immune responses appear to be indicative of increased prophylactic efficacy. Production and expression of several cytokines (interleukin 2 [IL-2], interferon γ, IL-12α, IL-1β, IL-6, and IL-22) were up-regulated in vaccinated animals. Human correlate studies revealed Sm-p80 reactivity with immunoglobulin G in human serum samples from schistosome-infected individuals. In addition, a complete lack of prevailing Sm-p80-specific immunoglobulin E in a high-risk or infected population was observed, thus minimizing the risk of hypersensitivity reaction following vaccination with Sm-p80 in humans. This study provided the proof of concept to move Sm-p80 forward into further preclinical development leading to human clinical trials.
Collapse
Affiliation(s)
- Gul Ahmad
- Department of Microbiology and Immunology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Bethony JM, Cole RN, Guo X, Kamhawi S, Lightowlers MW, Loukas A, Petri W, Reed S, Valenzuela JG, Hotez PJ. Vaccines to combat the neglected tropical diseases. Immunol Rev 2011; 239:237-70. [PMID: 21198676 DOI: 10.1111/j.1600-065x.2010.00976.x] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The neglected tropical diseases (NTDs) represent a group of parasitic and related infectious diseases such as amebiasis, Chagas disease, cysticercosis, echinococcosis, hookworm, leishmaniasis, and schistosomiasis. Together, these conditions are considered the most common infections in low- and middle-income countries, where they produce a level of global disability and human suffering equivalent to better known conditions such as human immunodeficiency virus/acquired immunodeficiency syndrome and malaria. Despite their global public health importance, progress on developing vaccines for NTD pathogens has lagged because of some key technical hurdles and the fact that these infections occur almost exclusively in the world's poorest people living below the World Bank poverty line. In the absence of financial incentives for new products, the multinational pharmaceutical companies have not embarked on substantive research and development programs for the neglected tropical disease vaccines. Here, we review the current status of scientific and technical progress in the development of new neglected tropical disease vaccines, highlighting the successes that have been achieved (cysticercosis and echinococcosis) and identifying the challenges and opportunities for development of new vaccines for NTDs. Also highlighted are the contributions being made by non-profit product development partnerships that are working to overcome some of the economic challenges in vaccine manufacture, clinical testing, and global access.
Collapse
Affiliation(s)
- Jeffrey M Bethony
- Microbiology, Immunology, and Tropical Medicine, George Washington University Medical Center, Washington, DC 20037, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Jørgensen LVG, Buchmann K. Cysteine proteases as potential antigens in antiparasitic DNA vaccines. Vaccine 2011; 29:5575-83. [PMID: 21664399 DOI: 10.1016/j.vaccine.2011.05.091] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 05/12/2011] [Accepted: 05/25/2011] [Indexed: 12/17/2022]
Abstract
Cysteine proteases in parasites are potent inducers of vertebrate host immune responses and may under certain circumstances take part in the pathogen's immune evasion strategies. These capacities place these parasite molecules as interesting candidate antigens in antiparasitic vaccines for use in vertebrates. Parasite cysteine proteases are able to skew the Th1/Th2 profile in mammals towards a response which allows sustainable parasite burdens in the host. DNA vaccines are also able to skew the Th1/Th2 profile by different administration techniques and the use of cysteine proteases in these genetic immunizations open perspectives for manipulation of the host immune response towards higher protection.
Collapse
Affiliation(s)
- Louise von Gersdorff Jørgensen
- Laboratory of Aquatic Pathobiology, Department of Veterinary Disease Biology, Faculty of Life Sciences, University of Copenhagen, Denmark.
| | | |
Collapse
|
48
|
Schistosoma mansoni antigen Sm-p80: Prophylactic efficacy of a vaccine formulated in human approved plasmid vector and adjuvant (VR 1020 and alum). Acta Trop 2011; 118:142-51. [PMID: 21334302 DOI: 10.1016/j.actatropica.2011.01.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Revised: 01/28/2011] [Accepted: 01/31/2011] [Indexed: 12/18/2022]
Abstract
Schistosomiasis is an important parasitic disease. Consensus is developing now that ideal control methods of the disease should be based on an integrated approach incorporating drug treatment, sanitation improvement, education, and an effective vaccine. With regards to the vaccine development, Sm-p80 has been shown to be a promising and strong immunogenic vaccine candidate. In the present study, Sm-p80-based vaccine formulated in alum was tested for its prophylactic efficacy in a mouse model. It was observed that vaccination using heterologous prime boost (DNA prime followed by boost with protein formulated in alum) and homologous prime boost (both prime and boost with protein formulated in alum) approaches, resulted in 61% and 55% reduction in worm burden, respectively. The protection was directly correlated with the induction of high titers of antibody responses that mainly included IgG, its isotypes, and IgM. In addition, both of the immunization approaches triggered a mixed Th1 and Th2 type response. Some involvement of Th17 specific immune response was also detected as indicated by the up-regulation of relevant cytokines. These results reinforce the potential of Sm-p80 as a viable vaccine candidate.
Collapse
|
49
|
Torben W, Ahmad G, Zhang W, Siddiqui AA. Role of antibodies in Sm-p80-mediated protection against Schistosoma mansoni challenge infection in murine and nonhuman primate models. Vaccine 2011; 29:2262-71. [PMID: 21277404 DOI: 10.1016/j.vaccine.2011.01.040] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Revised: 12/23/2010] [Accepted: 01/13/2011] [Indexed: 01/23/2023]
Abstract
Schistosomiasis is an important public health concern in more than 76 developing countries. Advent of an anti-schistosome vaccine would undoubtedly add to the existing control measures and may eventually help in the elimination of this disease. In the present study we have attempted to dissect the role(s) of antibodies in Sm-p80 mediated protection by intravenously transferring pooled sera from mice immunized with Sm-p80-pcDNA3 or purified IgG from baboons immunized with Sm-p80-pcDNA3, into naïve C57BL/6 mice, respectively, prior to challenge with cercariae. The passive transfer of antibodies from protected mice (homologous transfers) as well as transfer of total IgG from baboons (heterologous transfers), into naïve mice showed statistically significant reductions in worm burden and in the number of eggs in the tissues. Immunizations of antibody knockout mice (μMt-/-; B10.129S2 (B6)-Igh-6(tm1Cgn)/J) with recombinant Sm-p80 in the presence of CpG-motif oligodeoxynucleotides as an adjuvant, resulted in substantial reduction of Sm-p80-mediated protection, compared to C57BL/6 (normal) control group of mice. Down regulation of cytokines that have important effects on B cell proliferation as well as the recovery of higher number of parasites in antibody knockout indicated a significant role(s) of antibodies in Sm-p80-mediated protection against Schistosoma mansoni in mice. In toto, these studies appear to suggest that antibodies play a significant role in Sm-p80 mediated protection.
Collapse
Affiliation(s)
- Workineh Torben
- Department of Microbiology and Immunology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | | | | |
Collapse
|
50
|
Bergquist R, Lustigman S. Control of important helminthic infections vaccine development as part of the solution. ADVANCES IN PARASITOLOGY 2010; 73:297-326. [PMID: 20627146 DOI: 10.1016/s0065-308x(10)73010-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Among the tools available for the control of helminth infections, chemotherapy has come to totally dominate the field. In the veterinary field, development of drug resistance has appeared but this is not (yet) a problem in the control of human diseases. Although there is no vaccine commercially available for any human parasitic infection yet, recent progress in vaccine development is making this a future possibility for several diseases. The goal of chemotherapy is to alleviate infection and morbidity in the definitive host, or reduce transmission, while the effect of available vaccine candidates would mainly be to influence transmission through targeting the intermediate or reservoir host, when the infection is zoonotic. Apart from this general scheme, there are also vaccine candidates targeting the parasites in the definitive host, in particular the early developmental stages, which should reduce the risk of drug failure. Since the biological targets in most cases are different, vaccination would be synergistic with drug therapy. This review covers diseases caused by helminthes in both humans and animals and includes examples of diseases caused by cestodes, nematodes and trematodes. The focus is on infections for which vaccine development has been undertaken for a long time, resulting in products that could realistically become integrated into control strategies in the near future.
Collapse
|