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
|
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
|
3
|
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
|
4
|
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
|
5
|
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
|
6
|
Vaccination against the digestive enzyme Cathepsin B using a YS1646 Salmonella enterica Typhimurium vector provides almost complete protection against Schistosoma mansoni challenge in a mouse model. PLoS Negl Trop Dis 2019; 13:e0007490. [PMID: 31790394 PMCID: PMC6907844 DOI: 10.1371/journal.pntd.0007490] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 12/12/2019] [Accepted: 10/01/2019] [Indexed: 12/03/2022] Open
Abstract
Schistosoma mansoni threatens hundreds of millions of people in >50 countries. Schistosomulae migrate through the lung and adult worms reside in blood vessels adjacent to the intestinal mucosa. Current candidate vaccines aren’t designed to elicit a mucosal response. We have repurposed an attenuated Salmonella enterica Typhimurium strain (YS1646) to produce such a vaccine targeting Cathepsin B (CatB), a digestive enzyme important for parasite survival. Promoter-Type 3 secretory signal pairs were screened for protein expression in vitro and transfected into YS1646 to generate candidate vaccine strains. Two strains were selected for in vivo evaluation (nirB_SspH1 and SspH1_SspH1). Female C57BL/6 mice were immunized twice, 3 weeks apart, using six strategies: i) saline gavage (control), ii) the ‘empty’ YS1646 vector orally (PO) followed by intramuscular (IM) recombinant CatB (20μg IM rCatB), iii) two doses of IM rCatB, iv) two PO doses of YS1646-CatB, v) IM rCatB then PO YS1646-CatB and vi) PO YS1646-CatB then IM rCatB. Serum IgG responses to CatB were monitored by ELISA. Three weeks after the second dose, mice were challenged with 150 cercariae and sacrificed 7 weeks later to assess adult worm and egg burden (liver and intestine), granuloma size and egg morphology. CatB-specific IgG antibodies were low/absent in the control and PO only groups but rose substantially in other groups (5898-6766ng/mL). The highest response was in animals that received nirB_SspH1 YS1646 PO then IM rCatB. In this group, reductions in worm and intestine/liver egg burden (vs. control) were 93.1% and 79.5%/90.3% respectively (all P < .0001). Granuloma size was reduced in all vaccinated groups (range 32.9–52.8 x103μm2) and most significantly in the nirB_SspH1 + CatB IM group (34.7±3.4 x103μm2vs. 62.2±6.1 x103μm2: vs. control P < .01). Many eggs in the vaccinated animals had abnormal morphology. Targeting CatB using a multi-modality approach can provide almost complete protection against S. mansoni challenge. Schistosomiasis is a parasitic disease that affects over 250 million people worldwide and over 800 million are at risk of infection. Of the three main species, Schistosoma mansoni is the most widely distributed and is endemic in the Caribbean, South America, and Africa. It causes a chronic disease with severe negative effects on quality of life. Mass drug administration of praziquantel is the only available course of action due to a current lack of vaccines. However, praziquantel does not protect from reinfection. Therefore, a vaccine would be beneficial as a long-term solution to reduce morbidity and transmission of the disease. Our group has repurposed the attenuated YS1646 strain of Salmonella Typhimurium as an oral vaccine vector for the digestive enzyme Cathepsin B of S. mansoni. Oral vaccination followed by an intramuscular dose of recombinant Cathepsin B lead to significant reductions in parasite burden in mice. These animals had the highest titers in serum IgG and intestinal IgA antibodies. This multimodal vaccination approach also elicited both Th1 and Th2 cytokines as seen by the increases in IFNγ and IL-5. Finally, vaccinated mice had reductions in granuloma size along with a higher proportion of morphologically-abnormal eggs. This work demonstrates that a YS1646-based, multimodality, prime-boost immunization schedule can provide nearly complete protection against S. mansoni in a well-established murine model.
Collapse
|
7
|
Chaimon S, Limpanont Y, Reamtong O, Ampawong S, Phuphisut O, Chusongsang P, Ruangsittichai J, Boonyuen U, Watthanakulpanich D, O'Donoghue AJ, Caffrey CR, Adisakwattana P. Molecular characterization and functional analysis of the Schistosoma mekongi Ca 2+-dependent cysteine protease (calpain). Parasit Vectors 2019; 12:383. [PMID: 31362766 PMCID: PMC6668146 DOI: 10.1186/s13071-019-3639-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/25/2019] [Indexed: 11/22/2022] Open
Abstract
Background Schistosoma mekongi, which causes schistosomiasis in humans, is an important public health issue in Southeast Asia. Treatment with praziquantel is the primary method of control but emergence of praziquantel resistance requires the development of alternative drugs and vaccines. Calcium-dependent cysteine protease (calpain) is a novel vaccine candidate that has been studied in S. mansoni, S. japonicum, and protozoans including malaria, leishmania and trypanosomes. However, limited information is available on the properties and functions of calpain in other Schistosoma spp., including S. mekongi. In this study, we functionally characterized calpain 1 of S. mekongi (SmeCalp1). Results Calpain 1 of S. mekongi was obtained from transcriptomic analysis of S. mekongi; it had the highest expression level of all isoforms tested and was predominantly expressed in the adult male. SmeCalp1 cDNA is 2274 bp long and encodes 758 amino acids, with 85% to 90% homology with calpains in other Schistosoma species. Recombinant SmeCalp1 (rSmeCalp1), with a molecular weight of approximately 86.7 kDa, was expressed in bacteria and stimulated a marked antibody response in mice. Native SmeCalp1 was detected in crude worm extract and excretory-secretory product, and it was mainly localized in the tegument of the adult male; less signal was detected in the adult female worm. Thus, SmeCalp1 may play a role in surface membrane synthesis or host–parasite interaction. We assessed the protease activity of rSmeCalp1 and demonstrated that rSmeCalp1 could cleave the calpain substrate N-succinyl-Leu-Leu-Val-Tyr-7-amino-4-methylcoumarin, that was inhibited by calpain inhibitors (MDL28170 and E64c). Additionally, rSmeCalp1 could degrade the biological substrates fibronectin (blood clotting protein) and human complement C3, indicating important roles in the intravascular system and in host immune evasion. Conclusions SmeCalp1 is expressed on the tegumental surface of the parasite and can cleave host defense molecules; thus, it might participate in growth, development and survival during the entire life-cycle of S. mekongi. Information on the properties and functions of SmeCalp1 reported herein will be advantageous in the development of effective drugs and vaccines against S. mekongi and other schistosomes. Electronic supplementary material The online version of this article (10.1186/s13071-019-3639-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Salisa Chaimon
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Yanin Limpanont
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Orawan Phuphisut
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Phiraphol Chusongsang
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Jiraporn Ruangsittichai
- Department of Medical Entomology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Usa Boonyuen
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Dorn Watthanakulpanich
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Anthony J O'Donoghue
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, San Diego, California, USA
| | - Conor R Caffrey
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, San Diego, California, USA
| | - Poom Adisakwattana
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand.
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
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
|
15
|
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
|
16
|
Nawaratna SSK, Gobert GN, Willis C, Mulvenna J, Hofmann A, McManus DP, Jones MK. Lysosome-associated membrane glycoprotein (LAMP)--preliminary study on a hidden antigen target for vaccination against schistosomiasis. Sci Rep 2015; 5:15069. [PMID: 26472258 PMCID: PMC4607944 DOI: 10.1038/srep15069] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 08/13/2015] [Indexed: 12/12/2022] Open
Abstract
Our previously reported gene atlasing of schistosome tissues revealed transcripts that were highly enriched in the digestive tract of Schistosoma mansoni. From these, we selected two candidates, Sm-LAMP and Sm-NPC2 for testing as vaccine targets. The two molecules were selected on the basis of relatively high expression in the gastrodermis, their potentially important biological function, divergence from homologous molecules of the host and possible apical membrane expression in the gastrodermis. Bacterially expressed recombinant peptides corresponding to regions excluding trans-membrane domains of the selected vaccine targets were used in blinded vaccine trials in CBA mice using alum-CpG as adjuvant. Vaccine trials using the recombinant insoluble Sm-LAMP protein showed 16-25% significant reduction in total worm burden. Faecal egg count reduction was 52% and 60% in two trials, respectively, with similar results for the solubly expressed protein. Liver egg burden was reduced significantly (20% and 38%) with an insoluble recombinant Sm-LAMP in two trials, but not with the soluble recombinant form. Parasite fecundity was not affected by either Sm-LAMP protein preparations in the trials. It is concluded that Sm-LAMP may provide limited protection towards S. mansoni infections but could be used in combination with other vaccine candidates, to provide more comprehensive protection.
Collapse
Affiliation(s)
- Sujeevi S. K. Nawaratna
- School of Veterinary Sciences, The University of Queensland, Gatton Campus, Gatton Qld, 4343, Australia
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Qld, 4006, Australia
| | - Geoffrey N. Gobert
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Qld, 4006, Australia
| | - Charlene Willis
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Qld, 4006, Australia
| | - Jason Mulvenna
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Qld, 4006, Australia
| | - Andreas Hofmann
- Structural Chemistry Program, Eskitis Institute, Griffith University, Brisbane, Qld 4111, Australia
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Donald P. McManus
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Qld, 4006, Australia
| | - Malcolm K. Jones
- School of Veterinary Sciences, The University of Queensland, Gatton Campus, Gatton Qld, 4343, Australia
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Qld, 4006, Australia
| |
Collapse
|
17
|
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
|
18
|
Gonçalves de Assis NR, Batistoni de Morais S, Figueiredo BCP, Ricci ND, de Almeida LA, da Silva Pinheiro C, Martins VDP, Oliveira SC. DNA Vaccine Encoding the Chimeric Form of Schistosoma mansoni Sm-TSP2 and Sm29 Confers Partial Protection against Challenge Infection. PLoS One 2015; 10:e0125075. [PMID: 25942636 PMCID: PMC4420270 DOI: 10.1371/journal.pone.0125075] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/19/2015] [Indexed: 12/23/2022] Open
Abstract
Schistosomiasis is an important parasitic disease worldwide that affects more than 207 million people in 76 countries and causes approximately 250,000 deaths per year. The best long-term strategy to control schistosomiasis is through immunization combined with drug treatment. Due to the ability of DNA vaccines to generate humoral and cellular immune responses, such vaccines are considered a promising approach against schistosomiasis. Sm29 and tetraspanin-2 (Sm-TSP2) are two proteins that are located in the S. mansoni tegument of adult worms and schistosomula and induce high levels of protection through recombinant protein immunization. In this study, we transfected BHK-21 cells with plasmids encoding Sm29, Sm-TSP2 or a chimera containing both genes. Using RT-PCR analysis and western blot, we confirmed that the DNA vaccine constructs were transcribed and translated, respectively, in BHK-21 cells. After immunization of mice, we evaluated the reduction in worm burden. We observed worm burden reductions of 17-22%, 22%, 31-32% and 24-32% in animals immunized with the pUMVC3/Sm29, pUMVC3/SmTSP-2, pUMVC3/Chimera and pUMVC3/Sm29 + pUMVC3/SmTSP-2 plasmids, respectively. We evaluated the humoral response elicited by DNA vaccines, and animals immunized with pUMVC3/Sm29 and pUMVC3/Sm29 + pUMVC3/SmTSP-2 showed higher titers of anti-Sm29 antibodies. The cytokine profile produced by the spleen cells of immunized mice was then evaluated. We observed higher production of Th1 cytokines, such as TNF-α and IFN-γ, in vaccinated mice and no significant production of IL-4 and IL-5. The DNA vaccines tested in this study showed the ability to generate a protective immune response against schistosomiasis, probably through the production of Th1 cytokines. However, future strategies aiming to optimize the protective response induced by a chimeric DNA construct need to be developed.
Collapse
Affiliation(s)
- Natan Raimundo Gonçalves de Assis
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270–901, Belo Horizonte, MG, Brazil
| | - Suellen Batistoni de Morais
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270–901, Belo Horizonte, MG, Brazil
| | - Bárbara Castro Pimentel Figueiredo
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270–901, Belo Horizonte, MG, Brazil
| | - Natasha Delaqua Ricci
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270–901, Belo Horizonte, MG, Brazil
| | - Leonardo Augusto de Almeida
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
| | - Carina da Silva Pinheiro
- Departamento de Biointeração do Instituto de Ciências da Saúde, Universidade Federal da Bahia, 40110–100, Salvador, BA, Brazil
| | | | - Sergio Costa Oliveira
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270–901, Belo Horizonte, MG, Brazil
- * E-mail:
| |
Collapse
|
19
|
Mbanefo EC, Kumagai T, Kodama Y, Kurosaki T, Furushima-Shimogawara R, Cherif MS, Mizukami S, Kikuchi M, Huy NT, Ohta N, Sasaki H, Hirayama K. Immunogenicity and anti-fecundity effect of nanoparticle coated glutathione S-transferase (SjGST) DNA vaccine against murine Schistosoma japonicum infection. Parasitol Int 2015; 64:24-31. [PMID: 25603531 DOI: 10.1016/j.parint.2015.01.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 12/10/2014] [Accepted: 01/11/2015] [Indexed: 01/10/2023]
Abstract
There is still urgent need for a vaccine against schistosomiasis, especially in Schistosoma japonicum endemic areas where even a vaccine that will interrupt zoonotic transmission will be potentially effective as an intervention tool. We had developed a novel nanoparticle gene delivery system, which has proven efficacious in gene transfection to target immune cells with complementary adjuvant effect and high protective efficacy in several diseases. Here, we applied this nanoparticle system in combination with S. japonicum glutathione S-transferase (SjGST) DNA vaccine to show the immunogenicity and anti-fecundity effect of the nanoparticle coated vaccine formulation against murine schistosomiasis. The nanoparticle-coated DNA vaccine formulation induced desired immune responses. In comparison with the nanoparticle coated empty vector, it produced significantly increased antigen-specific humoral response, T-helper 1 polarized cytokine environment, higher proportion of IFN-γ producing CD4(+) T-cells and the concomitant decrease in IL-4 producing CD4(+) T-cells. Although there was no effect on worm burden, we recorded a marked reduction in tissue egg burden. There was up to 71.3% decrease in tissue egg burden and 55% reduction in the fecundity of female adult worms. Our data showed that SjGST DNA vaccine, delivered using the nanoparticle gene delivery system, produced anti-fecundity effect on female adult schistosomes as previously described by using conventional subunit vaccine with adjuvant, proving this DNA vaccine formulation as a promising candidate for anti-pathology and transmission blocking application.
Collapse
Affiliation(s)
- Evaristus Chibunna Mbanefo
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN) and Global COE Program, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Department of Parasitology and Entomology, Faculty of Bioscience, Nnamdi Azikiwe University, P.M.B. 5025, Awka, Nigeria
| | - Takashi Kumagai
- Section of Environmental Parasitology, Tokyo Medical and Dental University Graduate School of Medical and Dental Science, 113-8519, Japan
| | - Yukinobu Kodama
- Department of Hospital Pharmacy, Nagasaki University Hospital, 1-7-1 Sakamoto, 852-8501, Japan
| | - Tomoaki Kurosaki
- Department of Hospital Pharmacy, Nagasaki University Hospital, 1-7-1 Sakamoto, 852-8501, Japan
| | - Rieko Furushima-Shimogawara
- Section of Environmental Parasitology, Tokyo Medical and Dental University Graduate School of Medical and Dental Science, 113-8519, Japan
| | - Mahamoud Sama Cherif
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN) and Global COE Program, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan
| | - Shusaku Mizukami
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN) and Global COE Program, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan
| | - Mihoko Kikuchi
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN) and Global COE Program, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan
| | - Nguyen Tien Huy
- Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Department of Clinical Product Development, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan
| | - Nobuo Ohta
- Section of Environmental Parasitology, Tokyo Medical and Dental University Graduate School of Medical and Dental Science, 113-8519, Japan
| | - Hitoshi Sasaki
- Department of Hospital Pharmacy, Nagasaki University Hospital, 1-7-1 Sakamoto, 852-8501, Japan
| | - Kenji Hirayama
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN) and Global COE Program, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan.
| |
Collapse
|
20
|
Abstract
Helminth parasites infect over one fourth of the human population and are highly prevalent in livestock worldwide. In model systems, parasites are strongly immunomodulatory, but the immune system can be driven to expel them by prior vaccination. However, no vaccines are currently available for human use. Recent advances in vaccination with recombinant helminth antigens have been successful against cestode infections of livestock and new vaccines are being tested against nematode parasites of animals. Numerous vaccine antigens are being defined for a wide range of helminth parasite species, but greater understanding is needed to define the mechanisms of vaccine-induced immunity, to lay a rational platform for new vaccines and their optimal design. With human trials underway for hookworm and schistosomiasis vaccines, a greater integration between veterinary and human studies will highlight the common molecular and mechanistic pathways, and accelerate progress towards reducing the global health burden of helminth infection.
Collapse
|
21
|
Mbanefo EC, Huy NT, Wadagni AA, Eneanya CI, Nwaorgu O, Hirayama K. Host determinants of reinfection with schistosomes in humans: a systematic review and meta-analysis. PLoS Negl Trop Dis 2014; 8:e3164. [PMID: 25211227 PMCID: PMC4161334 DOI: 10.1371/journal.pntd.0003164] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 08/04/2014] [Indexed: 12/12/2022] Open
Abstract
Background Schistosomiasis is still a major public health burden in the tropics and subtropics. Although there is an effective chemotherapy (Praziquantel) for this disease, reinfection occurs rapidly after mass drug administration (MDA). Because the entire population do not get reinfected at the same rate, it is possible that host factors may play a dominant role in determining resistance or susceptibility to reinfection with schistosomes. Here, we systematically reviewed and meta-analyzed studies that reported associations between reinfection with the principal human-infecting species (S. mansoni, S. japonicum and S. haematobium) and host socio-demographic, epidemiological, immunological and genetic factors. Methodology/Principal Findings PubMed, Scopus, Google Scholar, Cochrane Review Library and African Journals Online public databases were searched in October 2013 to retrieve studies assessing association of host factors with reinfection with schistosomes. Meta-analysis was performed to generate pooled odds ratios and standardized mean differences as overall effect estimates for dichotomous and continuous variables, respectively. Quality assessment of included studies, heterogeneity between studies and publication bias were also assessed. Out of the initial 2739 records, 109 studies were included in the analyses, of which only 32 studies with 37 data sets were eligible for quantitative data synthesis. Among several host factors identified, strong positive association was found with age and pre-treatment intensity, and only slightly for gender. These factors are major determinants of exposure and disease transmission. Significant positive association was found with anti-SWA IgG4 level, and a negative overall effect for association with IgE levels. This reconfirmed the concept that IgE/IgG4 balance is a major determinant of protective immunity against schistosomiasis. Other identified determinants were reported by a small number of studies to enable interpretation. Conclusions Our data contribute to the understanding of host-parasite interaction as it affects reinfection, and is a potential tool to guide planning and tailoring of community interventions to target high-risk groups. One of the major challenges of schistosomiasis control is that disease prevalence reverts to baseline levels after mass drug administration due to high rate of reinfection. Host factors play a major role in determining resistance or susceptibility to reinfection with schistosomiasis and other diseases. We systematically searched and analyzed studies that identified potential host determinants of reinfection with schistosomes. Among demographic variables, age but not gender was strongly associated with reinfection with schistosomes. Pretreatment infection intensity was also identified as a major determinant of reinfection. Positive association with IgG4 levels and negative association with IgE levels reconfirmed the notion that IgE/IgG4 balance is the major factor controlling protective immunity against schistosomiasis. Other factors were reported by few studies to allow correct inferences. These results contribute to our understanding of host-parasite relationship as it affects reinfection, and will be useful for planning and targeting the limited resources for intervention on high-risk groups.
Collapse
Affiliation(s)
- Evaristus Chibunna Mbanefo
- Department of Parasitology and Entomology, Faculty of Bioscience, Nnamdi Azikiwe University, Awka, Nigeria
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
- * E-mail: ,
| | - Nguyen Tien Huy
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Anita Akpeedje Wadagni
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Christine Ifeoma Eneanya
- Department of Parasitology and Entomology, Faculty of Bioscience, Nnamdi Azikiwe University, Awka, Nigeria
| | - Obioma Nwaorgu
- Department of Parasitology and Entomology, Faculty of Bioscience, Nnamdi Azikiwe University, Awka, Nigeria
| | - Kenji Hirayama
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
| |
Collapse
|
22
|
Bahgat MM. Interaction Between the Neglected Tropical Disease Human Schistosomiasis and HCV Infection in Egypt: a Puzzling Relationship. J Clin Transl Hepatol 2014; 2:134-9. [PMID: 26356794 PMCID: PMC4521266 DOI: 10.14218/jcth.2013.00028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 02/21/2014] [Accepted: 02/26/2014] [Indexed: 02/06/2023] Open
Abstract
Egypt has the highest prevalence of chronic hepatitis C virus (HCV) infection and seropositivity worldwide, and it has been proposed that this enhanced susceptibility to HCV is related to coinfection with schistosomiasis. Although currently, there are no studies regarding the actual prevalence of both human schistosomiasis and schistosomiasis/HCV coinfection evidences strongly support that eliminating human schistosomiasis from Egypt is necessary to reduce both HCV prevalence and liver pathology. The present review highlights the significant impact of the neglected tropical disease human schistosomiasis on both susceptibility of Egyptians to HCV coinfection, severity of the resulting liver pathology, and poor response to antiviral therapy. The immune evasion mechanisms exerted by the HCV-NS3/4A protease domain, and the possible impact of immune evasion mechanisms exerted by proteases of larval, worm and egg stages of the parasite Schistosoma on human susceptibility to HCV infection are discussed. In addition, schistosome immune evasion mechanisms may include immunosuppression that in turn prevents clearance of HCV viremia and leads to relapsing HCV infection and severe liver pathology. I propose the generation of a replicon system from the most prevailing genotype (HCV-4a) in Egypt and establishing its replication on hepatoplastoma or immune cells in presence of bilharzial antigens. Finally, the use of a humanized small animal model that can acquire both HCV and S. mansoni infections will be important to further understand in real time the impact of coinfection on both the immune system and liver pathology.
Collapse
Affiliation(s)
- Mahmoud M. Bahgat
- Immunology and Infectious Diseases Group, Therapeutic Chemistry Department, the Centre of Excellence for Advanced Sciences, the National Research Centre, Dokki, Cairo, Egypt
- Research Group of Biomarkers for Infection and Immunity, Institute of Experimental Infection Research, TWINCORE Centre for Experimental and Clinical Infection Research, Hannover, Germany
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
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
|
26
|
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
|
27
|
Vaccination with enzymatically cleaved GPI-anchored proteins from Schistosoma mansoni induces protection against challenge infection. Clin Dev Immunol 2012; 2012:962538. [PMID: 22927873 PMCID: PMC3426240 DOI: 10.1155/2012/962538] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 06/21/2012] [Indexed: 12/05/2022]
Abstract
The flatworm Schistosoma mansoni is a blood fluke parasite that causes schistosomiasis, a debilitating disease that occurs throughout the developing world. Current schistosomiasis control strategies are mainly based on chemotherapy, but many researchers believe that the best long-term strategy to control schistosomiasis is through immunization with an antischistosomiasis vaccine combined with drug treatment. In the search for potential vaccine candidates, numerous tegument antigens have been assessed. As the major interface between parasite and mammalian host, the tegument plays crucial roles in the establishment and further course of schistosomiasis. Herein, we evaluated the potential of a GPI fraction, containing representative molecules located on the outer surface of adult worms, as vaccine candidate. Immunization of mice with GPI-anchored proteins induced a mixed Th1/Th2 type of immune response with production of IFN-γ and TNF-α, and low levels of IL-5 into the supernatant of splenocyte cultures. The protection engendered by this vaccination protocol was confirmed by 42% reduction in worm burden, 45% reduction in eggs per gram of hepatic tissue, 29% reduction in the number of granulomas per area, and 53% reduction in the granuloma fibrosis. Taken together, the data herein support the potential of surface-exposed GPI-anchored antigens from the S. mansoni tegument as vaccine candidate.
Collapse
|
28
|
Zhou Y, Zhang H, Sun XJ, Zheng D, Liang YJ, Luo J, Wang Y, Zhang ZS. Murine CD8+T cell cytotoxicity against schistosomula induced by inoculation of schistosomal 22.6/26GST coupled Sepharose 4B beads. Vaccine 2012; 30:2440-7. [DOI: 10.1016/j.vaccine.2012.01.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 01/16/2012] [Accepted: 01/21/2012] [Indexed: 10/14/2022]
|
29
|
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
|
30
|
Computational vaccinology: an important strategy to discover new potential S. mansoni vaccine candidates. J Biomed Biotechnol 2011; 2011:503068. [PMID: 22013383 PMCID: PMC3196198 DOI: 10.1155/2011/503068] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 08/12/2011] [Indexed: 11/17/2022] Open
Abstract
The flatworm Schistosoma mansoni is a blood fluke parasite that causes schistosomiasis, a debilitating disease that occurs throughout the developing world. Current schistosomiasis control strategies are mainly based on chemotherapy, but many researchers believe that the best long-term strategy to control schistosomiasis is through immunization with an antischistosomiasis vaccine combined with drug treatment. Several papers on Schistosoma mansoni vaccine and drug development have been published in the past few years, representing an important field of study. The advent of technologies that allow large-scale studies of genes and proteins had a remarkable impact on the screening of new and potential vaccine candidates in schistosomiasis. In this postgenomic scenario, bioinformatic technologies have emerged as important tools to mine transcriptomic, genomic, and proteomic databases. These new perspectives are leading to a new round of rational vaccine development. Herein, we discuss different strategies to identify potential S. mansoni vaccine candidates using computational vaccinology.
Collapse
|
31
|
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
|
32
|
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
|
33
|
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
|
34
|
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
|
35
|
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
|
36
|
Ahmad G, Zhang W, Torben W, Noor Z, Siddiqui AA. Protective effects of Sm-p80 in the presence of resiquimod as an adjuvant against challenge infection with Schistosoma mansoni in mice. Int J Infect Dis 2010; 14:e781-7. [PMID: 20630783 DOI: 10.1016/j.ijid.2010.02.2266] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 02/27/2010] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVES To determine the prophylactic efficacy of an Sm-p80-based vaccine formulation against challenge infection with Schistosoma mansoni in mice using an approach comprising of initial priming with DNA and boosting with recombinant protein in the presence of resiquimod (R848) as an adjuvant. METHODS In the first experiment (prime-boost approach), mice were primed with Sm-p80-pcDNA3 (week 0) and boosted at weeks 4 and 8 with recombinant Sm-p80 formulated in resiquimod (R848). Each mouse in the control group first received only pcDNA3 and was boosted with R848. In the second set of experiments (recombinant protein approach), mice were immunized (week 0) and boosted (weeks 4 and 8) with rSm-p80 formulated in R848. Animals of the control group in this series of experiments received only R848 at 0, 4, and 8 weeks. All of the animals from both the 'prime-boost' and 'recombinant protein' groups were challenged with cercariae of S. mansoni, 4 weeks after the last immunization. The mice were sacrificed 6 weeks post-challenge and the reductions in worm burden and egg production were determined. Sm-p80-specific antibody titers were estimated in the mice sera by ELISA. Cytokine mRNA and protein production by proliferating splenocytes in response to in vitro stimulation with Sm-p80, were estimated via RT-PCR and ELISA, respectively. RESULTS Vaccination with Sm-p80 (prime-boost approach) showed 49% reduction in worm burden; with the recombinant protein approach the protection was found to be 50%. The protection levels were correlated with antibody production. Upon antigenic stimulation with recombinant Sm-p80, splenocytes secreted significant levels of interferon (IFN)-γ and interleukin (IL)-2, indicating that the immune responses were Th1-biased and this was further supported in terms of distribution of antibody isotypes and mRNA expression of cytokines. CONCLUSIONS In conclusion the present study clearly demonstrates that Sm-p80 consistently maintained its protective nature, and resiquimod as an immunopotentiating agent slightly boosted the protective effects of Sm-p80 in both 'DNA prime-protein boost' and 'recombinant protein' immunization approaches in a murine model.
Collapse
Affiliation(s)
- Gul Ahmad
- Department of Microbiology and Immunology, Texas Tech University Health Sciences Center, 3601 4th Street, Mail Stop 6591, Lubbock, TX 79430, USA
| | | | | | | | | |
Collapse
|
37
|
Zhang W, Ahmad G, Torben W, Siddiqui AA. Sm-p80-based DNA vaccine made in a human use approved vector VR1020 protects against challenge infection with Schistosoma mansoni in mouse. Parasite Immunol 2010; 32:252-8. [PMID: 20398225 DOI: 10.1111/j.1365-3024.2009.01181.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Although there is an effective drug (praziquantel) available for the treatment of schistosomiasis, yet the disease is still spreading unabated and is rampant in 76 countries. Control via praziquantel treatment has so far been insufficient in reducing the disease transmission. Therefore, a vaccine in addition to other strategies, for example, improving sanitation and introduction of new drugs are essential to successfully control and eventually eradicate schistosomiasis. To this effect, we have targeted a functionally important antigen, Sm-p80 as a vaccine candidate. In this study, full length cDNA of Sm-p80 was cloned in VR1020, a FDA approved vector for human use. The protective efficacy of this vaccine formulation was tested in a murine model. Sm-p80-VR1020 vaccine formulation was able to induce 47% reduction in worm burden. Serology on samples obtained from vaccinated animals revealed a strong antibody response which included IgG and all of its subtypes, IgM and IgA. Proliferating splenocytes in response to recombinant Sm-p80 produced a wide spectrum of cytokines representing Th1, Th2 and Th17 types, as ascertained via RT-PCR analysis. These findings further strengthen the importance of Sm-p80 molecule as a vaccine candidate for intestinal schistosomiasis.
Collapse
Affiliation(s)
- W Zhang
- Department of Microbiology and Immunology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | | | | |
Collapse
|
38
|
Zhang W, Ahmad G, Torben W, Noor Z, Le L, Damian RT, Wolf RF, White GL, Chavez-Suarez M, Podesta RB, Kennedy RC, Siddiqui AA. Sm-p80-based DNA vaccine provides baboons with levels of protection against Schistosoma mansoni infection comparable to those achieved by the irradiated cercarial vaccine. J Infect Dis 2010; 201:1105-12. [PMID: 20187746 DOI: 10.1086/651147] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
To date, no vaccine is available to prevent human schistosomiasis. We have targeted a protein of Schistosoma mansoni that plays an important role in the surface membrane renewal process, a mechanism widely believed to be utilized by the parasite as an immune evasion strategy. Sm-p80 antigen is a promising vaccine target because of its documented immunogenicity, protective efficacy, and antifecundity effects observed in both experimental murine and nonhuman primate models of this infectious disease. In the present study, we report that, in a vector approved for human use (VR1020), an Sm-p80-based DNA vaccine formulation confers a 46% reduction in the worm burden in a baboon (Papio anubis) model. Baboons vaccinated with Sm-p80-VR1020 had a 28% decrease in egg production after challenge with the infectious parasite. Sm-p80-VR1020 vaccine elicited robust immune responses to specific antigen Sm-p80, including immunoglobulin (Ig) G, its subtypes IgG1 and IgG2, and IgA and IgM in vaccinated animals. When stimulated in vitro with recombinant Sm-p80, peripheral blood mononuclear cells and splenocytes from baboons vaccinated with Sm-p80-VR1020 produced considerably higher levels of T helper 1 response-enhancing cytokines (interleukin [IL]-2 and interferon-gamma) than T helper 2 (Th2) response-enhancing cytokines (IL-4 and IL-10). Peripheral blood mononuclear cells produced a significantly higher number of spot-forming units for interferon-gamma than for IL-4 in enzyme-linked immunosorbent spot assays. A mixed T helper 1/T helper 2 type of humoral and T cell responses was generated after immunization with Sm-p80-VR1020. These findings again highlight the potential of Sm-p80 as a promising vaccine candidate for schistosomiasis.
Collapse
Affiliation(s)
- Weidong Zhang
- Department of 1Microbiology and Immunology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ahmad G, Zhang W, Torben W, Haskins C, Diggs S, Noor Z, Le L, Siddiqui AA. Prime-boost and recombinant protein vaccination strategies using Sm-p80 protects against Schistosoma mansoni infection in the mouse model to levels previously attainable only by the irradiated cercarial vaccine. Parasitol Res 2009; 105:1767-77. [PMID: 19809833 DOI: 10.1007/s00436-009-1646-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Accepted: 09/23/2009] [Indexed: 12/17/2022]
Abstract
Advent of an effective schistosome vaccine would contribute significantly toward reducing the disease spectrum and transmission of schistosomiasis. We have targeted a functionally important antigen, Sm-p80, as a vaccine candidate because of its consistent immunogenicity, protective and antifecundity potentials, and important role in the immune evasion process. In this study, we report that using two vaccination approaches (prime boost and recombinant protein), Sm-p80-based vaccine formulation(s) confer up to 70% reduction in worm burden in mice. Animals immunized with the vaccine exhibited a decrease in egg production by up to 75%. The vaccine elicited strong immune responses that included IgM, IgA, and IgG (IgG1, IgG2a, IgG2b, and IgG3) in vaccinated animals. Splenocytes proliferated in response to Sm-p80 produced Th1 and Th17 response enhancing cytokines. These results again emphasize the potential of Sm-p80 as a viable vaccine candidate for schistosomiasis.
Collapse
Affiliation(s)
- Gul Ahmad
- Department of Microbiology and Immunology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Ahmad G, Torben W, Zhang W, Wyatt M, Siddiqui AA. Sm-p80-based DNA vaccine formulation induces potent protective immunity against Schistosoma mansoni. Parasite Immunol 2009; 31:156-61. [PMID: 19222788 DOI: 10.1111/j.1365-3024.2008.01091.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
No effective vaccine exists for the human parasitic disease, schistosomiasis. We have targeted a functionally important antigen, Sm-p80 as a vaccine candidate because of its consistent immunogenicity, protective potential and important role in the immune evasion process. In this study we report that a Sm-p80-based DNA vaccine formulation confers 59% reduction in worm burden in mice. Animals immunized with Sm-p80-pcDNA3 exhibited a decrease in egg production by 84%. Sm-p80 DNA elicited strong immune responses that include IgG2A and IgG2B antibody isotypes in vaccinated animals. Splenocytes proliferated in response to Sm-p80 produced appreciably more Th1 response enhancing cytokines (IL-2, IFN-gamma) than Th2 response enhancing cytokines (IL-4, IL-10). These data reinforce the potential of Sm-p80 as an excellent vaccine candidate for schistosomiasis.
Collapse
Affiliation(s)
- G Ahmad
- Department of Microbiology and Immunology, Texas Tech University Health Sciences Center, Lubbock, 79430, USA
| | | | | | | | | |
Collapse
|
41
|
Ahmad G, Zhang W, Torben W, Damian RT, Wolf RF, White GL, Chavez-Suarez M, Kennedy RC, Siddiqui AA. Protective and antifecundity effects of Sm-p80-based DNA vaccine formulation against Schistosoma mansoni in a nonhuman primate model. Vaccine 2009; 27:2830-7. [PMID: 19366570 DOI: 10.1016/j.vaccine.2009.02.096] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Revised: 02/19/2009] [Accepted: 02/24/2009] [Indexed: 12/26/2022]
Abstract
Schistosomiasis is an important parasitic disease for which there is no available vaccine. We have focused on a functionally important antigen of Schistosoma mansoni, Sm-p80, as a vaccine candidate because of its consistent immunogenicity, protective potential and antifecundity effect observed in murine models; and for its pivotal role in the immune evasion process. In the present study we report that an Sm-p80-based DNA vaccine formulation confers 38% reduction in worm burden in a nonhuman primate model, the baboon (Papio anubis). Animals immunized with Sm-p80-pcDNA3 exhibited a decrease in egg production by 32%. Sm-p80 DNA elicited specific immune responses that include IgG; its subtypes IgG1 and IgG2; and IgM in vaccinated animals. Peripheral blood mononuclear cells (PBMCs) from immunized animals when stimulated in vitro with Sm-p80 produced appreciably more Th1 response enhancing cytokines (IL-2, IFN-gamma) than Th2 response enhancing cytokines (IL-4, IL-10). PBMCs produced appreciably more spot-forming units for INF-gamma than for IL-4 in enzyme-linked immunosorbent spot (ELISPOT) assays. Overall it appears that even though a mixed (Th1/Th2) type of humoral antibody response was generated following immunization with Sm-p80; the dominant protective immune response is Th1 type. These data reinforce the potential of Sm-p80 as an excellent vaccine candidate for schistosomiasis.
Collapse
Affiliation(s)
- Gul Ahmad
- Department of Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States
| | | | | | | | | | | | | | | | | |
Collapse
|