1
|
Koff WC, Rappuoli R, Plotkin SA. Historical advances in structural and molecular biology and how they impacted vaccine development. J Mol Biol 2023; 435:168113. [PMID: 37080423 DOI: 10.1016/j.jmb.2023.168113] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/11/2023] [Accepted: 04/11/2023] [Indexed: 04/22/2023]
Abstract
Vaccines are among the greatest tools for prevention and control of disease. They have eliminated smallpox from the planet, decreased morbidity and mortality for major infectious diseases like polio, measles, mumps, and rubella, significantly blunted the impact of the COVID-19 pandemic, and prevented viral induced cancers such as cervical cancer caused by human papillomavirus. Recent technological advances, in genomics, structural biology, and human immunology have transformed vaccine development, enabling new technologies such as mRNA vaccines to greatly accelerate development of new and improved vaccines. In this review, we briefly highlight the history of vaccine development, and provide examples of where advances in genomics and structural biology, paved the way for development of vaccines for bacterial and viral diseases.
Collapse
Affiliation(s)
- Wayne C Koff
- President and CEO, Human Immunome Project, New York, NY, USA
| | - Rino Rappuoli
- Chief Scientific Officer, Fondazione Biotechnopolo, Siena, Italy
| | - Stanley A Plotkin
- Emeritus Professor of Pediatrics, University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
2
|
Khalid F, Tahir R, Ellahi M, Amir N, Rizvi SFA, Hasnain A. Emerging trends of edible vaccine therapy for combating human diseases especially
COVID
‐19: Pros, cons, and future challenges. Phytother Res 2022; 36:2746-2766. [PMID: 35499291 PMCID: PMC9347755 DOI: 10.1002/ptr.7475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/18/2022] [Accepted: 04/06/2022] [Indexed: 11/07/2022]
Abstract
The researchers are still doing efforts to develop an effective, reliable, and easily accessible vaccine candidate to protect against COVID‐19. As of the August 2020, nearly 30 conventional vaccines have been emerged in clinical trials, and more than 200 vaccines are in various development stages. Nowadays, plants are also considered as a potential source for the production of monoclonal antibodies, vaccines, drugs, immunomodulatory proteins, as well as used as bioreactors or factories for their bulk production. The scientific evidences enlighten that plants are the rich source of oral vaccines, which can be given either by eating the edible parts of plants and/or by oral administration of highly refined proteins. The use of plant‐based edible vaccines is an emerging trend as it possesses minimum or no side effects compared with synthetic vaccines. This review article gives insights into different types of vaccines, the use of edible vaccines, advantages of edible vaccines over conventional vaccines, and mechanism of action of edible vaccines. This review article also focuses on the applications of edible vaccines in wide‐range of human diseases especially against COVID‐19 with emphasis on future perspectives of the use of edible vaccines.
Collapse
Affiliation(s)
- Fatima Khalid
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
| | - Reema Tahir
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
| | - Manahil Ellahi
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
| | - Nilofer Amir
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
| | - Syed Faheem Askari Rizvi
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
- College of Chemistry and Chemical EngineeringLanzhou UniversityLanzhouP.R. China
| | - Ammarah Hasnain
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
| |
Collapse
|
3
|
Kalinina AA, Nesterenko LN, Bruter AV, Balunets DV, Chudakov DM, Izraelson M, Britanova OV, Khromykh LM, Kazansky DB. Adoptive Immunotherapy Based on Chain-Centric TCRs in Treatment of Infectious Diseases. iScience 2020; 23:101854. [PMID: 33313494 PMCID: PMC7721641 DOI: 10.1016/j.isci.2020.101854] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/03/2020] [Accepted: 11/19/2020] [Indexed: 12/29/2022] Open
Abstract
Complications after vaccination, lack of vaccines against certain infections, and the emergence of antibiotic-resistant microorganisms point to the need for alternative ways of protection and treatment of infectious diseases. Here, we proposed a therapeutic approach to control salmonellosis based on adoptive cell therapy. We showed that the T cell receptor (TCR) repertoire of salmonella-specific memory cells contains 20% of TCR variants with the dominant-active α-chain. Transduction of intact T lymphocytes with the dominant salmonella-specific TCRα led to their enhanced in vitro proliferation in response to salmonella. Adoptive transfer of transduced T cells resulted in a significant decrease in bacterial loads in mice infected with salmonella before or after the adoptive transfer. We demonstrated that adoptive immunotherapy based on T cells, transduced with dominant-specific TCRα could be successfully applied for treatment and prevention of infectious diseases and represent a useful addition to vaccination and existing therapeutic strategies. A regular TCR repertoire of memory T cells contains alpha-chain-centric TCRs Dominant-active TCRα, paired with random TCRβ, recognizes specific microbial antigens Adoptive immunotherapy could be applied for treatment of infections
Collapse
Affiliation(s)
- Anastasiia A Kalinina
- Federal State Budgetary Institution "N. N. Blokhin National Medical Research Center of Oncology" оf the Ministry of Health of the Russian Federation, 115478 Moscow, Russia
| | - Ludmila N Nesterenko
- "N. F. Gamaleya National Research Center of Epidemiology and Microbiology", the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Alexandra V Bruter
- Federal State Budgetary Institution "N. N. Blokhin National Medical Research Center of Oncology" оf the Ministry of Health of the Russian Federation, 115478 Moscow, Russia.,Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Denis V Balunets
- "N. F. Gamaleya National Research Center of Epidemiology and Microbiology", the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Dmitriy M Chudakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Mark Izraelson
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Olga V Britanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Ludmila M Khromykh
- Federal State Budgetary Institution "N. N. Blokhin National Medical Research Center of Oncology" оf the Ministry of Health of the Russian Federation, 115478 Moscow, Russia
| | - Dmitry B Kazansky
- Federal State Budgetary Institution "N. N. Blokhin National Medical Research Center of Oncology" оf the Ministry of Health of the Russian Federation, 115478 Moscow, Russia
| |
Collapse
|
4
|
Optimal protection against Salmonella infection requires noncirculating memory. Proc Natl Acad Sci U S A 2018; 115:10416-10421. [PMID: 30254173 DOI: 10.1073/pnas.1808339115] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
While CD4 Th1 cells are required for resistance to intramacrophage infections, adoptive transfer of Th1 cells is insufficient to protect against Salmonella infection. Using an epitope-tagged vaccine strain of Salmonella, we found that effective protection correlated with expanded Salmonella-specific memory CD4 T cells in circulation and nonlymphoid tissues. However, naive mice that previously shared a blood supply with vaccinated partners lacked T cell memory with characteristics of tissue residence and did not acquire robust protective immunity. Using a YFP-IFN-γ reporter system, we identified Th1 cells in the liver of immunized mice that displayed markers of tissue residence, including P2X7, ARTC2, LFA-1, and CD101. Adoptive transfer of liver memory cells after ARTC2 blockade increased protection against highly virulent bacteria. Taken together, these data demonstrate that noncirculating memory Th1 cells are a vital component of immunity to Salmonella infection and should be the focus of vaccine strategies.
Collapse
|
5
|
Breurec S, Rafaï C, Onambele M, Frank T, Farra A, Legrand A, Weill FX. Serotype Distribution and Antimicrobial Resistance of Shigella Species in Bangui, Central African Republic, from 2002 to 2013. Am J Trop Med Hyg 2018; 99:283-286. [PMID: 29943713 DOI: 10.4269/ajtmh.17-0917] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Shigella is a major cause of severe diarrhea in children less than the age of 5 years in sub-Saharan Africa. The aim of this study was to describe the (sub-)serotype distribution and antimicrobial susceptibility of Shigella serogroups from Centrafrican patients with diarrhea between 2002 and 2013. We collected 443 Shigella isolates in total. The most common serogroups were Shigella flexneri (N = 243, 54.9%), followed by Shigella sonnei (N = 90, 20.3%) and Shigella dysenteriae (N = 72, 16.3%). The high diversity of (sub-)serotypes of S. flexneri and S. dysenteriae may impede the development of an efficient vaccine. Rates of resistance were high for ampicillin, chloramphenicol, tetracycline, and cotrimoxazole but low for many other antimicrobials, confirming recommendations for the use of third-generation cephalosporins (only one organism resistant) and fluoroquinolones (no resistance). However, the detection of one extended-spectrum beta-lactamase-producing Shigella organism highlights the need for continued monitoring of antimicrobial drug susceptibility.
Collapse
Affiliation(s)
- Sebastien Breurec
- Laboratoire de Bactériologie, Institut Pasteur de Bangui, Bangui, Central African Republic.,Laboratoire de Microbiologie Clinique et Environnementale, Centre Hospitalier Universitaire de Pointe-à-Pitre/les Abymes, Pointe-à-Pitre, France.,Faculté de Médecine Hyacinthe Bastaraud, Université des Antilles, Pointe-à-Pitre, France
| | - Clotaire Rafaï
- Laboratoire de Bactériologie, Institut Pasteur de Bangui, Bangui, Central African Republic
| | - Manuella Onambele
- Laboratoire de Bactériologie, Institut Pasteur de Bangui, Bangui, Central African Republic
| | - Thierry Frank
- Laboratoire de Bactériologie, Institut Pasteur de Bangui, Bangui, Central African Republic
| | - Alain Farra
- Laboratoire de Bactériologie, Institut Pasteur de Bangui, Bangui, Central African Republic
| | - Arnaud Legrand
- Délégation à la Recherche Clinique et à l'Innovation, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - François-Xavier Weill
- Unité des Bactéries Pathogènes Entériques, Centre National de Référence des Escherichia coli, Shigella et Salmonella, Institut Pasteur, Paris, France
| |
Collapse
|
6
|
Abstract
French Guiana, a tropical country, is characterised by a young and multi-ethnic population. Difficulties in accessing safe water sources lead to outbreaks of gastroenteritis. The objectives of this study were (1) to describe the microbiological profile of shigella strains isolated in western French Guiana, including antimicrobial susceptibility and the distribution of strains in terms of species and serotypes and (2) to estimate the incidence of shigellosis in children under 5 years old. A retrospective observational study was conducted of 213 cases of shigellosis diagnosed in the biology department of the hospital centre for western French Guiana between 2000 and 2012 in children under 5 years old. The serogroups (formerly known as species) that predominates in French Guiana was Shigella flexneri. No resistance was observed to fluoroquinolones or to third-generation cephalosporins. The average incidence of shigellosis in children under 5 years old in western French Guiana was estimated at 189.6 cases per 100 000 inhabitants per year. Shigellosis is a public health problem in western French Guiana. These infections suggest the difficulties in accessing safe water sources and the lack of public sanitation. A quadrivalent vaccine containing Shigella sonnei and three serotypes of S. flexneri (S. flexneri 2a, 3a and 6) could provide broad coverage against shigella infections.
Collapse
|
7
|
Lee SJ, Benoun J, Sheridan BS, Fogassy Z, Pham O, Pham QM, Puddington L, McSorley SJ. Dual Immunization with SseB/Flagellin Provides Enhanced Protection against Salmonella Infection Mediated by Circulating Memory Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:1353-1361. [PMID: 28710253 PMCID: PMC5548602 DOI: 10.4049/jimmunol.1601357] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 06/19/2017] [Indexed: 01/09/2023]
Abstract
The development of a subunit Salmonella vaccine has been hindered by the absence of detailed information about antigenic targets of protective Salmonella-specific T and B cells. Recent studies have identified SseB as a modestly protective Ag in susceptible C57BL/6 mice, but the mechanism of protective immunity remains undefined. In this article, we report that simply combining Salmonella SseB with flagellin substantially enhances protective immunity, allowing immunized C57BL/6 mice to survive for up to 30 d following challenge with virulent bacteria. Surprisingly, the enhancing effect of flagellin did not require flagellin Ag targeting during secondary responses or recognition of flagellin by TLR5. Although coimmunization with flagellin did not affect SseB-specific Ab responses, it modestly boosted CD4 responses. In addition, protective immunity was effectively transferred in circulation to parabionts of immunized mice, demonstrating that tissue-resident memory is not required for vaccine-induced protection. Finally, protective immunity required host expression of IFN-γR but was independent of induced NO synthase expression. Taken together, these data indicate that Salmonella flagellin has unique adjuvant properties that improve SseB-mediated protective immunity provided by circulating memory.
Collapse
Affiliation(s)
- Seung-Joo Lee
- Center for Comparative Medicine, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616; and
| | - Joseph Benoun
- Center for Comparative Medicine, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616; and
| | - Brian S Sheridan
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Zachary Fogassy
- Center for Comparative Medicine, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616; and
| | - Oanh Pham
- Center for Comparative Medicine, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616; and
| | - Quynh-Mai Pham
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Lynn Puddington
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Stephen J McSorley
- Center for Comparative Medicine, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616;
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616; and
| |
Collapse
|
8
|
Felgner J, Jain A, Nakajima R, Liang L, Jasinskas A, Gotuzzo E, Vinetz JM, Miyajima F, Pirmohamed M, Hassan-Hanga F, Umoru D, Jibir BW, Gambo S, Olateju K, Felgner PL, Obaro S, Davies DH. Development of ELISAs for diagnosis of acute typhoid fever in Nigerian children. PLoS Negl Trop Dis 2017. [PMID: 28640809 PMCID: PMC5498068 DOI: 10.1371/journal.pntd.0005679] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Improved serodiagnostic tests for typhoid fever (TF) are needed for surveillance, to facilitate patient management, curb antibiotic resistance, and inform public health programs. To address this need, IgA, IgM and IgG ELISAs using Salmonella enterica serovar Typhi (S. Typhi) lipopolysaccharide (LPS) and hemolysin E (t1477) protein were conducted on 86 Nigerian pediatric TF and 29 non-typhoidal Salmonella (NTS) cases, 178 culture-negative febrile cases, 28 "other" (i.e., non-Salmonella) pediatric infections, and 48 healthy Nigerian children. The best discrimination was achieved between TF and healthy children. LPS-specific IgA and IgM provided receiver operator characteristic areas under the curve (ROC AUC) values of 0.963 and 0.968, respectively, and 0.978 for IgA+M combined. Similar performance was achieved with t1477-specific IgA and IgM (0.968 and 0.968, respectively; 0.976 combined). IgG against LPS and t1477 was less accurate for discriminating these groups, possibly as a consequence of previous exposure, although ROC AUC values were still high (0.928 and 0.932, respectively). Importantly, discrimination between TF and children with other infections was maintained by LPS-specific IgA and IgM (AUC = 0.903 and 0.934, respectively; 0.938 combined), and slightly reduced for IgG (0.909), while t1477-specific IgG performed best (0.914). A similar pattern was seen when comparing TF with other infections from outside Nigeria. The t1477 may be recognized by cross-reactive antibodies from other acute infections, although a robust IgG response may provide some diagnostic utility in populations where incidence of other infections is low, such as in children. The data are consistent with IgA and IgM against S. Typhi LPS being specific markers of acute TF.
Collapse
Affiliation(s)
- Jiin Felgner
- Division of Infectious Diseases, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Aarti Jain
- Division of Infectious Diseases, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Rie Nakajima
- Division of Infectious Diseases, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Li Liang
- Division of Infectious Diseases, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Algis Jasinskas
- Division of Infectious Diseases, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Eduardo Gotuzzo
- Alexander von Humboldt Institute of Tropical Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
- Hospital Nacional Cayetano Heredia, Lima, Peru
| | - Joseph M. Vinetz
- Alexander von Humboldt Institute of Tropical Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
- Division of Infectious Diseases, Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Fabio Miyajima
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | - Munir Pirmohamed
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | | | | | | | - Safiya Gambo
- Department of Pediatrics, Murtala Specialist Hospital, Kano, Nigeria
| | | | - Philip L. Felgner
- Division of Infectious Diseases, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Stephen Obaro
- Department of Pediatrics, Aminu Kano Teaching Hospital, Kano, Nigeria
- University of Abuja Teaching Hospital, Gwagwalada, Nigeria
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- International Foundation against Infectious Diseases in Nigeria (IFAIN), Abuja, Nigeria
| | - D. Huw Davies
- Division of Infectious Diseases, School of Medicine, University of California Irvine, Irvine, California, United States of America
- * E-mail:
| |
Collapse
|
9
|
de Barros JMS, Costabile A, Charalampopoulos D, Khutoryanskiy VV, Edwards AD. Evaluating and optimizing oral formulations of live bacterial vaccines using a gastro-small intestine model. Eur J Pharm Biopharm 2016; 102:115-22. [PMID: 26969261 DOI: 10.1016/j.ejpb.2016.03.010] [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] [Received: 10/30/2015] [Revised: 02/27/2016] [Accepted: 03/07/2016] [Indexed: 10/22/2022]
Abstract
Gastrointestinal (GI) models that mimic physiological conditions in vitro are important tools for developing and optimizing biopharmaceutical formulations. Oral administration of live attenuated bacterial vaccines (LBV) can safely and effectively promote mucosal immunity but new formulations are required that provide controlled release of optimal numbers of viable bacterial cells, which must survive gastrointestinal transit overcoming various antimicrobial barriers. Here, we use a gastro-small intestine gut model of human GI conditions to study the survival and release kinetics of two oral LBV formulations: the licensed typhoid fever vaccine Vivotif comprising enteric coated capsules; and an experimental formulation of the model vaccine Salmonella Typhimurium SL3261 dried directly onto cast enteric polymer films and laminated to form a polymer film laminate (PFL). Neither formulation released significant numbers of viable cells when tested in the complete gastro-small intestine model. The poor performance in delivering viable cells could be attributed to a combination of acid and bile toxicity plus incomplete release of cells for Vivotif capsules, and to bile toxicity alone for PFL. To achieve effective protection from intestinal bile in addition to effective acid resistance, bile adsorbent resins were incorporated into the PFL to produce a new formulation, termed BR-PFL. Efficient and complete release of 4.4×10(7) live cells per dose was achieved from BR-PFL at distal intestinal pH, with release kinetics controlled by the composition of the enteric polymer film, and no loss in viability observed in any stage of the GI model. Use of this in vitro GI model thereby allowed rational design of an oral LBV formulation to maximize viable cell release.
Collapse
Affiliation(s)
- João M S de Barros
- School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD, UK
| | - Adele Costabile
- Food and Nutritional Sciences, University of Reading, Whiteknights, Reading RG6 6AD, UK
| | | | | | - Alexander D Edwards
- School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD, UK.
| |
Collapse
|
10
|
Leitner DR, Lichtenegger S, Temel P, Zingl FG, Ratzberger D, Roier S, Schild-Prüfert K, Feichter S, Reidl J, Schild S. A combined vaccine approach against Vibrio cholerae and ETEC based on outer membrane vesicles. Front Microbiol 2015; 6:823. [PMID: 26322032 PMCID: PMC4531250 DOI: 10.3389/fmicb.2015.00823] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/27/2015] [Indexed: 11/28/2022] Open
Abstract
Enteric infections induced by pathogens like Vibrio cholerae and enterotoxigenic Escherichia coli (ETEC) remain a massive burden in developing countries with increasing morbidity and mortality rates. Previously, we showed that the immunization with genetically detoxified outer membrane vesicles (OMVs) derived from V. cholerae elicits a protective immune response based on the generation of O antigen antibodies, which effectively block the motility by binding to the sheathed flagellum. In this study, we investigated the potential of lipopolysaccharide (LPS)-modified and toxin negative OMVs isolated from V. cholerae and ETEC as a combined OMV vaccine candidate. Our results indicate that the immunization with V. cholerae or ETEC OMVs induced a species-specific immune response, whereas the combination of both OMV species resulted in a high-titer, protective immune response against both pathogens. Interestingly, the immunization with V. cholerae OMVs alone resulted in a so far uncharacterized and cholera toxin B-subunit (CTB) independent protection mechanism against an ETEC colonization. Furthermore, we investigated the potential use of V. cholerae OMVs as delivery vehicles for the heterologously expression of the ETEC surface antigens, CFA/I, and FliC. Although we induced a detectable immune response against both heterologously expressed antigens, none of these approaches resulted in an improved protection compared to a simple combination of V. cholerae and ETEC OMVs. Finally, we expanded the current protection model from V. cholerae to ETEC by demonstrating that the inhibition of motility via anti-FliC antibodies represents a relevant protection mechanism of an OMV-based ETEC vaccine candidate in vivo.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Stefan Schild
- Institute of Molecular Biosciences, University of GrazGraz, Austria
| |
Collapse
|
11
|
Nag D, Sinha R, Mitra S, Barman S, Takeda Y, Shinoda S, Chakrabarti MK, Koley H. Heat killed multi-serotype Shigella immunogens induced humoral immunity and protection against heterologous challenge in rabbit model. Immunobiology 2015. [PMID: 26210044 DOI: 10.1016/j.imbio.2015.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Recently we have shown the homologous protective efficacy of heat killed multi-serotype Shigella (HKMS) immunogens in a guinea pig colitis model. In our present study, we have advanced our research by immunizing rabbits with a reduced number of oral doses and evaluating the host's adaptive immune responses. The duration of immunogenicity and subsequently protective efficacy was determined against wild type heterologous Shigella strains in a rabbit luminal model. After three successive oral immunizations with HKMS immunogens, serum and lymphocyte supernatant antibody titer against the heterologous shigellae were reciprocally increased and remained at an elevated level up to 180 days. Serogroup and serotype specific O-antigen of lipopolysaccharide and immunogenic proteins of heterologous challenge strains were detected by immunoblot assay. Up-regulation of IL-12p35, IFN-γ and IL-10 mRNA expression was detected in immunized rabbit peripheral blood mononuclear cells (PBMC) after stimulation with HKMS in vitro. HKMS-specific plasma cell response was confirmed by production of a relatively higher level of HKMS-specific IgG in immunized PBMC supernatant compared to control group. Furthermore, the immunized groups of rabbits exhibited complete protection against wild type heterologous shigellae challenge. Thus HKMS immunogens induced humoral and Th1-mediated adaptive immunity and provided complete protection in a rabbit model. These immunogens could be a broad spectrum non-living vaccine candidate for human use in the near future.
Collapse
Affiliation(s)
- Dhrubajyoti Nag
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Ritam Sinha
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Soma Mitra
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Soumik Barman
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Yoshifumi Takeda
- Collaborative Research Centre of Okayama University for Infectious Diseases, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Sumio Shinoda
- Collaborative Research Centre of Okayama University for Infectious Diseases, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - M K Chakrabarti
- Division of Pathophysiology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Hemanta Koley
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases, Kolkata, India.
| |
Collapse
|
12
|
Saleem AF, Mach O, Quadri F, Khan A, Bhatti Z, Rehman NU, Zaidi S, Weldon WC, Oberste SM, Salama M, Sutter RW, Zaidi AKM. Immunogenicity of poliovirus vaccines in chronically malnourished infants: a randomized controlled trial in Pakistan. Vaccine 2015; 33:2757-63. [PMID: 25917673 PMCID: PMC4447616 DOI: 10.1016/j.vaccine.2015.04.055] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 04/11/2015] [Accepted: 04/14/2015] [Indexed: 11/29/2022]
Abstract
Reaching high population immunity against polioviruses (PV) is essential to achieving global polio eradication. Efficacy of oral poliovirus vaccine (OPV) varies and is lower among children living in tropical areas with impoverished environments. Malnutrition found as a risk factor for lower serological protection against PV. We compared whether inactivated polio vaccine (IPV) can be used to rapidly close the immunity gap among chronically malnourished (stunted) infants in Pakistan who will not be eligible for the 14 week IPV dose in routine EPI schedule. A phase 3, multicenter 4-arm randomized controlled trial conducted at five Primary Health Care (PHC) centers in Karachi, Pakistan. Infants, 9–12 months were stratified by length for age Z score into chronically malnourished and normally nourished. Infants were randomized to receive one dose of either bivalent OPV (bOPV) alone or bOPV + IPV. Baseline seroprevalence of PV antibodies and serum immune response to study vaccine dose were assessed by neutralization assay. Vaccine PV shedding in stool was evaluated 7 days after a bOPV challenge dose. Sera and stool were analyzed from 852/928 (92%) enrolled children. At baseline, the seroprevalence was 85.6% (n = 386), 73.6% (n = 332), and 70.7% (n = 319) in malnourished children against PV types 1, 2 and 3 respectively; and 94.1% (n = 448), 87.0% (n = 441) and 83.6% (n = 397) in the normally nourished group (p < 0.05). Children had previously received 9–10 doses of bOPV (80%) or tOPV (20%). One dose of IPV + bOPV given to malnourished children increased their serological protection (PV1, n = 201, 97.6%; PV2, n = 198, 96.1% and PV3, n = 189, 91.7%) to parity with normally nourished children who had not received IPV (p = <0.001). Seroconversion and boosting for all three serotypes was significantly more frequent in children who received IPV + bOPV than in those with bOPV only (p < 0.001) in both strata. Shedding of polioviruses in stool did not differ between study groups and ranged from 2.4% (n = 5) to 7.1% (n = 15). In malnourished children the shedding was reduced after bOPV + IPV compared to bOPV only. Chronically malnourished infants were more likely to be unprotected against polioviruses than normal infants. bOPV + IPV helped close the immunity gap better than bOPV alone.
Collapse
Affiliation(s)
- Ali Faisal Saleem
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Ondrej Mach
- Polio Eradication Department, World Health Organization, Geneva, Switzerland
| | - Farheen Quadri
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Asia Khan
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Zaid Bhatti
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Najeeb Ur Rehman
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Sohail Zaidi
- Department of virology, National Institute of Health, Islamabad, Pakistan
| | - William C Weldon
- Population Immunity Laboratory, Polio and Picornavirus Laboratory Branch, Centers for Disease Control and Prevention, Atlanta, USA
| | - Steven M Oberste
- Polio and Picornavirus Laboratory Branch, Centers for Disease Control and Prevention, Atlanta, USA
| | | | - Roland W Sutter
- Polio Eradication Department, World Health Organization, Geneva, Switzerland
| | - Anita K M Zaidi
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan.
| |
Collapse
|
13
|
Anders KL, Thompson CN, Thuy NTV, Nguyet NM, Tu LTP, Dung TTN, Phat VV, Van NTH, Hieu NT, Tham NTH, Ha PTT, Lien LB, Chau NVV, Baker S, Simmons CP. The epidemiology and aetiology of diarrhoeal disease in infancy in southern Vietnam: a birth cohort study. Int J Infect Dis 2015; 35:3-10. [PMID: 25813553 PMCID: PMC4508461 DOI: 10.1016/j.ijid.2015.03.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/13/2015] [Accepted: 03/17/2015] [Indexed: 01/11/2023] Open
Abstract
The diarrhoeal disease burden in a large, prospective infant cohort in Vietnam is defined. Minimum incidence of clinic-based diarrhoea in infants: 271/1000 infant-years. Rotavirus was most commonly identified, followed by norovirus and bacterial pathogens. Frequent repeat infections with the same pathogen within 1 year. Inclusion of rotavirus in the immunization schedule for Vietnam is warranted.
Objectives Previous studies indicate a high burden of diarrhoeal disease in Vietnamese children, however longitudinal community-based data on burden and aetiology are limited. The findings from a large, prospective cohort study of diarrhoeal disease in infants in southern Vietnam are presented herein. Methods Infants were enrolled at birth in urban Ho Chi Minh City and a semi-rural district in southern Vietnam, and followed for 12 months (n = 6706). Diarrhoeal illness episodes were identified through clinic-based passive surveillance, hospital admissions, and self-reports. Results The minimum incidence of diarrhoeal illness in the first year of life was 271/1000 infant-years of observation for the whole cohort. Rotavirus was the most commonly detected pathogen (50% of positive samples), followed by norovirus (24%), Campylobacter (20%), Salmonella (18%), and Shigella (16%). Repeat infections were identified in 9% of infants infected with rotavirus, norovirus, Shigella, or Campylobacter, and 13% of those with Salmonella infections. Conclusions The minimum incidence of diarrhoeal disease in infants in both urban and semi-rural settings in southern Vietnam was quantified prospectively. A large proportion of laboratory-diagnosed disease was caused by rotavirus and norovirus. These data highlight the unmet need for a rotavirus vaccine in Vietnam and provide evidence of the previously unrecognized burden of norovirus in infants.
Collapse
Affiliation(s)
- Katherine L Anders
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hospital for Tropical Diseases, 764 Vo Van Kiet, District 5, Ho Chi Minh City, Vietnam; Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, Oxford University, Oxford, UK; Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia.
| | - Corinne N Thompson
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hospital for Tropical Diseases, 764 Vo Van Kiet, District 5, Ho Chi Minh City, Vietnam; Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, Oxford University, Oxford, UK; The London School of Hygiene and Tropical Medicine, London, UK
| | - Nguyen Thi Van Thuy
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hospital for Tropical Diseases, 764 Vo Van Kiet, District 5, Ho Chi Minh City, Vietnam
| | - Nguyen Minh Nguyet
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hospital for Tropical Diseases, 764 Vo Van Kiet, District 5, Ho Chi Minh City, Vietnam; The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Le Thi Phuong Tu
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hospital for Tropical Diseases, 764 Vo Van Kiet, District 5, Ho Chi Minh City, Vietnam
| | - Tran Thi Ngoc Dung
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hospital for Tropical Diseases, 764 Vo Van Kiet, District 5, Ho Chi Minh City, Vietnam
| | - Voong Vinh Phat
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hospital for Tropical Diseases, 764 Vo Van Kiet, District 5, Ho Chi Minh City, Vietnam
| | - Nguyen Thi Hong Van
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hospital for Tropical Diseases, 764 Vo Van Kiet, District 5, Ho Chi Minh City, Vietnam
| | | | | | | | - Le Bich Lien
- Children's Hospital No. 1, Ho Chi Minh City, Vietnam
| | | | - Stephen Baker
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hospital for Tropical Diseases, 764 Vo Van Kiet, District 5, Ho Chi Minh City, Vietnam; Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, Oxford University, Oxford, UK; The London School of Hygiene and Tropical Medicine, London, UK
| | - Cameron P Simmons
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hospital for Tropical Diseases, 764 Vo Van Kiet, District 5, Ho Chi Minh City, Vietnam; Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, Oxford University, Oxford, UK; Department of Microbiology and Immunology, University of Melbourne, Victoria, Australia
| |
Collapse
|
14
|
Salem W, Leitner DR, Zingl FG, Schratter G, Prassl R, Goessler W, Reidl J, Schild S. Antibacterial activity of silver and zinc nanoparticles against Vibrio cholerae and enterotoxic Escherichia coli. Int J Med Microbiol 2015; 305:85-95. [PMID: 25466205 PMCID: PMC4300426 DOI: 10.1016/j.ijmm.2014.11.005] [Citation(s) in RCA: 182] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 10/30/2014] [Accepted: 11/04/2014] [Indexed: 11/17/2022] Open
Abstract
Vibrio cholerae and enterotoxic Escherichia coli (ETEC) remain two dominant bacterial causes of severe secretory diarrhea and still a significant cause of death, especially in developing countries. In order to investigate new effective and inexpensive therapeutic approaches, we analyzed nanoparticles synthesized by a green approach using corresponding salt (silver or zinc nitrate) with aqueous extract of Caltropis procera fruit or leaves. We characterized the quantity and quality of nanoparticles by UV-visible wavelength scans and nanoparticle tracking analysis. Nanoparticles could be synthesized in reproducible yields of approximately 10(8) particles/ml with mode particles sizes of approx. 90-100 nm. Antibacterial activity against two pathogens was assessed by minimal inhibitory concentration assays and survival curves. Both pathogens exhibited similar resistance profiles with minimal inhibitory concentrations ranging between 5×10(5) and 10(7) particles/ml. Interestingly, zinc nanoparticles showed a slightly higher efficacy, but sublethal concentrations caused adverse effects and resulted in increased biofilm formation of V. cholerae. Using the expression levels of the outer membrane porin OmpT as an indicator for cAMP levels, our results suggest that zinc nanoparticles inhibit adenylyl cyclase activity. This consequently deceases the levels of this second messenger, which is a known inhibitor of biofilm formation. Finally, we demonstrated that a single oral administration of silver nanoparticles to infant mice colonized with V. cholerae or ETEC significantly reduces the colonization rates of the pathogens by 75- or 100-fold, respectively.
Collapse
Affiliation(s)
- Wesam Salem
- University of Graz, Institute of Molecular Biosciences, BioTechMed-Graz, Humboldtstrasse 50, A-8010 Graz, Austria; South Valley University, Faculty of Science, Qena, Egypt
| | - Deborah R Leitner
- University of Graz, Institute of Molecular Biosciences, BioTechMed-Graz, Humboldtstrasse 50, A-8010 Graz, Austria
| | - Franz G Zingl
- University of Graz, Institute of Molecular Biosciences, BioTechMed-Graz, Humboldtstrasse 50, A-8010 Graz, Austria
| | - Gebhart Schratter
- Institute of Biophysics, Medical University of Graz, BioTechMed-Graz, Schmiedlstraße 6, 8042 Graz, Austria
| | - Ruth Prassl
- Institute of Biophysics, Medical University of Graz, BioTechMed-Graz, Schmiedlstraße 6, 8042 Graz, Austria
| | - Walter Goessler
- Institute for Chemistry, Analytical Chemistry, University of Graz, BioTechMed-Graz, 8010 Graz, Austria
| | - Joachim Reidl
- University of Graz, Institute of Molecular Biosciences, BioTechMed-Graz, Humboldtstrasse 50, A-8010 Graz, Austria
| | - Stefan Schild
- University of Graz, Institute of Molecular Biosciences, BioTechMed-Graz, Humboldtstrasse 50, A-8010 Graz, Austria.
| |
Collapse
|
15
|
Tanner R, Kakalacheva K, Miller E, Pathan AA, Chalk R, Sander CR, Scriba T, Tameris M, Hawkridge T, Mahomed H, Hussey G, Hanekom W, Checkley A, McShane H, Fletcher HA. Serum indoleamine 2,3-dioxygenase activity is associated with reduced immunogenicity following vaccination with MVA85A. BMC Infect Dis 2014; 14:660. [PMID: 25466778 PMCID: PMC4265419 DOI: 10.1186/s12879-014-0660-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 11/24/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND There is an urgent need for improved vaccines to protect against tuberculosis. The currently available vaccine Bacille Calmette-Guerin (BCG) has varying immunogenicity and efficacy across different populations for reasons not clearly understood. MVA85A is a modified vaccinia virus expressing antigen 85A from Mycobacterium tuberculosis which has been in clinical development since 2002 as a candidate vaccine to boost BCG-induced protection. A recent efficacy trial in South African infants failed to demonstrate enhancement of protection over BCG alone. The immunogenicity was lower than that seen in UK trials. The enzyme Indoleamine 2,3-dioxygenase (IDO) catalyses the first and rate-limiting step in the breakdown of the essential amino acid tryptophan. T cells are dependent on tryptophan and IDO activity suppresses T-cell proliferation and function. METHODS Using samples collected during phase I trials with MVA85A across the UK and South Africa we have investigated the relationship between vaccine immunogenicity and IDO using IFN-γ ELISPOT, qPCR and liquid chromatography mass spectrometry. RESULTS We demonstrate an IFN-γ dependent increase in IDO mRNA expression in peripheral blood mononuclear cells (PBMC) following MVA85A vaccination in UK subjects. IDO mRNA correlates positively with the IFN-γ ELISPOT response indicating that vaccine specific induction of IDO in PBMC is unlikely to limit the development of vaccine specific immunity. IDO activity in the serum of volunteers from the UK and South Africa was also assessed. There was no change in serum IDO activity following MVA85A vaccination. However, we observed higher baseline IDO activity in South African volunteers when compared to UK volunteers. In both UK and South African serum samples, baseline IDO activity negatively correlated with vaccine-specific IFN-γ responses, suggesting that IDO activity may impair the generation of a CD4+ T cell memory response. CONCLUSIONS Baseline IDO activity was higher in South African volunteers when compared to UK volunteers, which may represent a potential mechanism for the observed variation in vaccine immunogenicity in South African and UK populations and may have important implications for future vaccination strategies. TRIAL REGISTRATION Trials are registered at ClinicalTrials.gov; UK cohort NCT00427830, UK LTBI cohort NCT00456183, South African cohort NCT00460590, South African LTBI cohort NCT00480558.
Collapse
Affiliation(s)
- Rachel Tanner
- The Jenner Institute, University of Oxford, Oxford, UK.
| | - Kristina Kakalacheva
- The Jenner Institute, University of Oxford, Oxford, UK. .,Present address: Department of Neuroinflammation, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| | - Ellen Miller
- The Jenner Institute, University of Oxford, Oxford, UK. .,Present address: Royal Sussex County Hospital, Eastern road, Brighton, UK.
| | - Ansar A Pathan
- The Jenner Institute, University of Oxford, Oxford, UK. .,Present address: Centre for Infection, Immunity and Disease Mechanisms, Biosciences, School of Health Sciences and Social Care, Brunel University, Middlesex, UK.
| | - Rod Chalk
- Structural Genomics Consortium, University of Oxford, Oxford, UK.
| | - Clare R Sander
- The Jenner Institute, University of Oxford, Oxford, UK. .,Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - Tom Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa.
| | - Michelle Tameris
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa.
| | - Tony Hawkridge
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa. .,Vaccines for Africa Initiative, Cape Town, South Africa.
| | - Hassan Mahomed
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa. .,Division of Community Health, Stellenbosch University, Stellenbosch, South Africa. .,Metropolitan District Health Services, Western Cape, Government: Health, Cape Town, South Africa.
| | - Greg Hussey
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa. .,Vaccines for Africa Initiative, Cape Town, South Africa.
| | - Willem Hanekom
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa.
| | - Anna Checkley
- The Jenner Institute, University of Oxford, Oxford, UK. .,Present address: London School of Hygiene and Tropical Medicine, Keppel Street, London, UK.
| | - Helen McShane
- The Jenner Institute, University of Oxford, Oxford, UK.
| | - Helen A Fletcher
- The Jenner Institute, University of Oxford, Oxford, UK. .,Present address: London School of Hygiene and Tropical Medicine, Keppel Street, London, UK.
| |
Collapse
|
16
|
Nanton MR, Lee SJ, Atif SM, Nuccio SP, Taylor JJ, Bäumler AJ, Way SS, McSorley SJ. Direct visualization of endogenous Salmonella-specific B cells reveals a marked delay in clonal expansion and germinal center development. Eur J Immunol 2014; 45:428-41. [PMID: 25346524 DOI: 10.1002/eji.201444540] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 09/30/2014] [Accepted: 10/21/2014] [Indexed: 11/11/2022]
Abstract
CD4(+) T cells and B cells are both essential for acquired immunity to Salmonella infection. It is well established that Salmonella inhibit host CD4(+) T-cell responses, but a corresponding inhibitory effect on B cells is less well defined. Here, we utilize an Ag tetramer and pull-down enrichment strategy to directly visualize OVA-specific B cells in mice, as they respond to infection with Salmonella-OVA. Surprisingly, OVA-specific B-cell expansion and germinal center formation was not detected until bacteria were cleared from the host. Furthermore, Salmonella infection also actively inhibited both B- and T-cell responses to the same coinjected Ag but this did not require the presence of iNOS. The Salmonella Pathogenicity Island 2 (SPI2) locus has been shown to be responsible for inhibition of Salmonella-specific CD4(+) T-cell responses, and an examination of SPI2-deficient bacteria demonstrated a recovery in B-cell expansion in infected mice. Together, these data suggest that Salmonella can simultaneously inhibit host B- and T-cell responses using SPI2-dependent mechanisms.
Collapse
Affiliation(s)
- Minelva R Nanton
- Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA; Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota Medical School-Twin Cities, Minneapolis, MN, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
de Barros JM, Scherer T, Charalampopoulos D, Khutoryanskiy VV, Edwards AD. A Laminated Polymer Film Formulation for Enteric Delivery of Live Vaccine and Probiotic Bacteria. J Pharm Sci 2014; 103:2022-2032. [DOI: 10.1002/jps.23997] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 03/04/2014] [Accepted: 04/09/2014] [Indexed: 01/12/2023]
|
18
|
Livio S, Strockbine NA, Panchalingam S, Tennant SM, Barry EM, Marohn ME, Antonio M, Hossain A, Mandomando I, Ochieng JB, Oundo JO, Qureshi S, Ramamurthy T, Tamboura B, Adegbola RA, Hossain MJ, Saha D, Sen S, Faruque ASG, Alonso PL, Breiman RF, Zaidi AKM, Sur D, Sow SO, Berkeley LY, O'Reilly CE, Mintz ED, Biswas K, Cohen D, Farag TH, Nasrin D, Wu Y, Blackwelder WC, Kotloff KL, Nataro JP, Levine MM. Shigella isolates from the global enteric multicenter study inform vaccine development. Clin Infect Dis 2014; 59:933-41. [PMID: 24958238 PMCID: PMC4166982 DOI: 10.1093/cid/ciu468] [Citation(s) in RCA: 264] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Shigella, a major diarrheal disease pathogen worldwide, is the target of vaccine development. The Global Enteric Multicenter Study (GEMS) investigated burden and etiology of moderate-to-severe diarrheal disease in children aged <60 months and matched controls without diarrhea during 3 years at 4 sites in Africa and 3 in Asia. Shigella was 1 of the 4 most common pathogens across sites and age strata. GEMS Shigella serotypes are reviewed to guide vaccine development. METHODS Subjects' stool specimens/rectal swabs were transported to site laboratories in transport media and plated onto xylose lysine desoxycholate and MacConkey agar. Suspect Shigella colonies were identified by biochemical tests and agglutination with antisera. Shigella isolates were shipped to the GEMS Reference Laboratory (Baltimore, MD) for confirmation and serotyping of S. flexneri; one-third of isolates were sent to the Centers for Disease Control and Prevention for quality control. RESULTS Shigella dysenteriae and S. boydii accounted for 5.0% and 5.4%, respectively, of 1130 Shigella case isolates; S. flexneri comprised 65.9% and S. sonnei 23.7%. Five serotypes/subserotypes comprised 89.4% of S. flexneri, including S. flexneri 2a, S. flexneri 6, S. flexneri 3a, S. flexneri 2b, and S. flexneri 1b. CONCLUSIONS A broad-spectrum Shigella vaccine must protect against S. sonnei and 15 S. flexneri serotypes/subserotypes. A quadrivalent vaccine with O antigens from S. sonnei, S. flexneri 2a, S. flexneri 3a, and S. flexneri 6 can provide broad direct coverage against these most common serotypes and indirect coverage against all but 1 (rare) remaining subserotype through shared S. flexneri group antigens.
Collapse
Affiliation(s)
- Sofie Livio
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | - Nancy A Strockbine
- Escherichia and Shigella Reference Unit, Enteric Diseases Laboratory Branch Division of Foodborne, Waterborne and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Sandra Panchalingam
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | - Sharon M Tennant
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | - Eileen M Barry
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | - Mark E Marohn
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | - Martin Antonio
- Medical Research Council Unit (United Kingdom), Fajara, The Gambia
| | - Anowar Hossain
- International Centre for Diarrhoeal Disease Research, Mohakhali, Dhaka, Bangladesh
| | - Inacio Mandomando
- Centro de Investigação em Saúde de Manhiça, Manhiça, Mozambique and the Centre de Recerca en Salut Internacional de Barcelona, Hospital Clinic/Universitat de Barcelona, Spain
| | - John B Ochieng
- Kenya Medical Research Institute/Centers for Disease Control and Prevention, Kisumu, Kenya
| | - Joseph O Oundo
- Kenya Medical Research Institute/Centers for Disease Control and Prevention, Kisumu, Kenya
| | - Shahida Qureshi
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | | | - Boubou Tamboura
- Centre pour le Développement des Vaccins du Mali, Bamako, Mali
| | | | | | - Debasish Saha
- Medical Research Council Unit (United Kingdom), Fajara, The Gambia
| | - Sunil Sen
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | | | - Pedro L Alonso
- Centro de Investigação em Saúde de Manhiça, Manhiça, Mozambique and the Centre de Recerca en Salut Internacional de Barcelona, Hospital Clinic/Universitat de Barcelona, Spain
| | - Robert F Breiman
- Kenya Medical Research Institute/Centers for Disease Control and Prevention, Kisumu, Kenya Global Disease Detection Division, Kenya Office of the US Centers for Disease Control and Prevention, Nairobi, Kenya
| | - Anita K M Zaidi
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Dipika Sur
- National Institute of Cholera and Enteric Diseases, Kolkata, India Program for Appropriate Technology in Health (PATH), New Delhi, India
| | - Samba O Sow
- Centre pour le Développement des Vaccins du Mali, Bamako, Mali
| | - Lynette Y Berkeley
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore US Food and Drug Administration, Rockville
| | - Ciara E O'Reilly
- Division of Foodborne, Waterborne and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Eric D Mintz
- Division of Foodborne, Waterborne and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Kousick Biswas
- Department of Veterans Affairs, Cooperative Studies Program Coordinating Center, Perry Point, Maryland
| | - Dani Cohen
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | - Tamer H Farag
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | - Dilruba Nasrin
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | - Yukun Wu
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | - William C Blackwelder
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | - Karen L Kotloff
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | - James P Nataro
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | - Myron M Levine
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| |
Collapse
|
19
|
Oh KH, Kim DW, Jung SM, Cho SH. Molecular characterization of Enterotoxigenic Escherichia coli strains isolated from diarrheal patients in Korea during 2003-2011. PLoS One 2014; 9:e96896. [PMID: 24841334 PMCID: PMC4026316 DOI: 10.1371/journal.pone.0096896] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 04/12/2014] [Indexed: 11/18/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is one of the major causes of infectious diarrhea in developing countries. In order to characterize the molecular features of human ETEC isolates from Korea, we investigated the profiles of enterotoxin and colonization factor (CF) genes by polymerase chain reaction (PCR) and performed multilocus sequence typing (MLST) with a total of 291 ETEC strains. The specimens comprised 258 domestic strains isolated from patients who had diarrhea and were from widely separated geographic regions in Korea and 33 inflow strains isolated from travelers visiting other Asian countries. Heat-stable toxin (STh)-possessing ETEC strains were more frequent than heat-labile toxin (LT)-possessing ETEC strains in the domestic isolates, while the detection rates of both enterotoxin genes were similar in the inflow isolates. The profile of CF genes of domestic isolates was similar to that of inflow isolates and the major CF types of the strains were CS3-CS21-CS1/PCF071 and CS2-CS3-CS21. Most of these 2 CF types were detected in ETEC strains that possess both lt and sth genes. The major MLSTST types of domestic isolates were ST171 and ST955. Moreover, the 2 major CF types were usually found concomitantly with the 2 major MLST STs, ST171 and ST955. In conclusion, our genotyping results may provide useful information for guiding the development of geographically specific vaccines against human ETEC isolates.
Collapse
Affiliation(s)
- Kyung-Hwan Oh
- Division of Enteric Bacterial Infections, Center for Infectious Diseases, Korea National Institute of Health, Osong-eup, Chungcheongbuk-do, Republic of Korea
| | - Dong Wook Kim
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Kyeonggi-do, Republic of Korea
| | - Su-Mi Jung
- Division of Enteric Bacterial Infections, Center for Infectious Diseases, Korea National Institute of Health, Osong-eup, Chungcheongbuk-do, Republic of Korea
| | - Seung-Hak Cho
- Division of Enteric Bacterial Infections, Center for Infectious Diseases, Korea National Institute of Health, Osong-eup, Chungcheongbuk-do, Republic of Korea
| |
Collapse
|
20
|
Heine SJ, Diaz-McNair J, Andar AU, Drachenberg CB, van de Verg L, Walker R, Picking WL, Pasetti MF. Intradermal delivery of Shigella IpaB and IpaD type III secretion proteins: kinetics of cell recruitment and antigen uptake, mucosal and systemic immunity, and protection across serotypes. THE JOURNAL OF IMMUNOLOGY 2014; 192:1630-40. [PMID: 24453241 DOI: 10.4049/jimmunol.1302743] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Shigella is one of the leading pathogens contributing to the vast pediatric diarrheal disease burden in low-income countries. No licensed vaccine is available, and the existing candidates are only partially effective and serotype specific. Shigella type III secretion system proteins IpaB and IpaD, which are conserved across Shigella spp., are candidates for a broadly protective, subunit-based vaccine. In this study, we investigated the immunogenicity and protective efficacy of IpaB and IpaD administered intradermally (i.d.) with a double-mutant of the Escherichia coli heat-labile enterotoxin (dmLT) adjuvant using microneedles. Different dosage levels of IpaB and IpaD, with or without dmLT, were tested in mice. Vaccine delivery into the dermis, recruitment of neutrophils, macrophages, dendritic cells, and Langerhans cells, and colocalization of vaccine Ag within skin-activated APC were demonstrated through histology and immunofluorescence microscopy. Ag-loaded neutrophils, macrophages, dendritic cells, and Langerhans cells remained in the tissue at least 1 wk. IpaB, IpaD, and dmLT-specific serum IgG- and IgG-secreting cells were produced following i.d. immunization. The protective efficacy was 70% against Shigella flexneri and 50% against Shigella sonnei. Similar results were obtained when the vaccine was administered intranasally, with the i.d. route requiring 25-40 times lower doses. Distinctively, IgG was detected in mucosal secretions; secretory IgA, as well as mucosal and systemic IgA Ab-secreting cells, were seemingly absent. Vaccine-induced T cells produced IFN-γ, IL-2, TNF-α, IL-17, IL-4, IL-5, and IL-10. These results demonstrate the potential of i.d. vaccination with IpaB and IpaD to prevent Shigella infection and support further studies in humans.
Collapse
Affiliation(s)
- Shannon J Heine
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Several live-attenuated Shigella vaccines, with well-defined mutations in specific genes, have shown great promise in eliciting significant immune responses when given orally to volunteers. These responses have been measured by evaluating antibody-secreting cells, serum antibody levels and fecal immunoglobulin A to bacterial lipopolysaccharide and to individual bacterial invasion plasmid antigens. In this review, data collected from volunteer trials with live Shigella vaccines from three different research groups are described. The attenuating features of the bacterial strains, as well as the immune response following the use of different dosing regimens, are also described. The responses obtained with each vaccine strain are compared with data obtained from challenge trials using wild-type Shigella strains. Although the exact correlates of protection have not been found, some consensus may be derived as to what may constitute a protective immune response. Future directions in the field of live Shigella vaccines are also discussed.
Collapse
Affiliation(s)
- Malabi M Venkatesan
- Division of Bacterial and Rickettsial Diseases, Walter Reed Army Institute of Research, 503 Robert Forney Drive, Room 3s12, Silver Spring, MD 20910, USA.
| | | |
Collapse
|
22
|
Concise synthesis of di- and trisaccharides related to the O-antigens from Shigella flexneri serotypes 6 and 6a, based on late stage mono-O-acetylation and/or site-selective oxidation. Tetrahedron 2013. [DOI: 10.1016/j.tet.2013.10.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
23
|
Ruane D, Brane L, Reis BS, Cheong C, Poles J, Do Y, Zhu H, Velinzon K, Choi JH, Studt N, Mayer L, Lavelle EC, Steinman RM, Mucida D, Mehandru S. Lung dendritic cells induce migration of protective T cells to the gastrointestinal tract. ACTA ACUST UNITED AC 2013; 210:1871-88. [PMID: 23960190 PMCID: PMC3754860 DOI: 10.1084/jem.20122762] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Developing efficacious vaccines against enteric diseases is a global challenge that requires a better understanding of cellular recruitment dynamics at the mucosal surfaces. The current paradigm of T cell homing to the gastrointestinal (GI) tract involves the induction of α4β7 and CCR9 by Peyer's patch and mesenteric lymph node (MLN) dendritic cells (DCs) in a retinoic acid-dependent manner. This paradigm, however, cannot be reconciled with reports of GI T cell responses after intranasal (i.n.) delivery of antigens that do not directly target the GI lymphoid tissue. To explore alternative pathways of cellular migration, we have investigated the ability of DCs from mucosal and nonmucosal tissues to recruit lymphocytes to the GI tract. Unexpectedly, we found that lung DCs, like CD103(+) MLN DCs, up-regulate the gut-homing integrin α4β7 in vitro and in vivo, and induce T cell migration to the GI tract in vivo. Consistent with a role for this pathway in generating mucosal immune responses, lung DC targeting by i.n. immunization induced protective immunity against enteric challenge with a highly pathogenic strain of Salmonella. The present report demonstrates novel functional evidence of mucosal cross talk mediated by DCs, which has the potential to inform the design of novel vaccines against mucosal pathogens.
Collapse
Affiliation(s)
- Darren Ruane
- Laboratory of Cellular Immunology and Physiology and 2 Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Levine MM, Kotloff KL, Nataro JP, Muhsen K. The Global Enteric Multicenter Study (GEMS): impetus, rationale, and genesis. Clin Infect Dis 2013; 55 Suppl 4:S215-24. [PMID: 23169934 PMCID: PMC3502311 DOI: 10.1093/cid/cis761] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Diarrheal disease remains one of the top 2 causes of young child mortality in the developing world. Whereas improvements in water/sanitation infrastructure and hygiene can diminish transmission of enteric pathogens, vaccines can also hasten the decline of diarrheal disease morbidity and mortality. From 1980 through approximately 2004, various case/control and small cohort studies were undertaken to address the etiology of pediatric diarrhea in developing countries. Many studies had methodological limitations and came to divergent conclusions, making it difficult to prioritize the relative importance of different pathogens. Consequently, in the first years of the millennium there was no consensus on what diarrheal disease vaccines should be developed or implemented; however, there was consensus on the need for a well-designed study to obtain information on the etiology and burden of more severe forms of diarrheal disease to guide global investment and implementation decisions. Accordingly, the Global Enteric Multicenter Study (GEMS) was designed to overcome drawbacks of earlier studies and determine the etiology and population-based burden of pediatric diarrheal disease. GEMS, which includes one of the largest case/control studies of an infectious disease syndrome ever undertaken (target approximately 12 600 analyzable cases and 12 600 controls), was rolled out in 4 sites in sub-Saharan Africa (Gambia, Kenya, Mali, Mozambique) and 3 in South Asia (Bangladesh, India, Pakistan), with each site linked to a population under demographic surveillance (total approximately 467 000 child years of observation among children <5 years of age). GEMS data will guide investment and help prioritize strategies to mitigate the morbidity and mortality of pediatric diarrheal disease.
Collapse
Affiliation(s)
- Myron M Levine
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | | | | | |
Collapse
|
25
|
Farag TH, Nasrin D, Wu Y, Muhsen K, Blackwelder WC, Sommerfelt H, Panchalingam S, Nataro JP, Kotloff KL, Levine MM. Some epidemiologic, clinical, microbiologic, and organizational assumptions that influenced the design and performance of the Global Enteric Multicenter Study (GEMS). Clin Infect Dis 2013; 55 Suppl 4:S225-31. [PMID: 23169935 PMCID: PMC3502315 DOI: 10.1093/cid/cis787] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The overall aim of the Global Enteric Multicenter Study–1 (GEMS-1) is to identify the etiologic agents associated with moderate-to-severe diarrhea (MSD) among children <5 years of age, and thereby the attributable pathogen-specific population-based incidence of MSD, to guide investments in research and public health interventions against diarrheal disease. To accomplish this, 9 core assumptions were vetted through widespread consultation: (1) a limited number of etiologic agents may be responsible for most MSD; (2) a definition of MSD can be crafted that encompasses cases that might otherwise be fatal in the community without treatment; (3) MSD seen at sentinel centers is a proxy for fatal diarrheal disease in the community; (4) matched case/control is the appropriate epidemiologic design; (5) methods across the sites can be standardized and rigorous quality control maintained; (6) a single 60-day postenrollment visit to case and control households creates mini-cohorts, allowing comparisons; (7) broad support for GEMS-1 messages can be achieved by incorporating advice from public health spokespersons; (8) results will facilitate the setting of investment and intervention priorities; and (9) wide acceptance and dissemination of the GEMS-1 results can be achieved.
Collapse
Affiliation(s)
- Tamer H Farag
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Blackwelder WC, Biswas K, Wu Y, Kotloff KL, Farag TH, Nasrin D, Graubard BI, Sommerfelt H, Levine MM. Statistical methods in the Global Enteric Multicenter Study (GEMS). Clin Infect Dis 2013; 55 Suppl 4:S246-53. [PMID: 23169937 PMCID: PMC3502316 DOI: 10.1093/cid/cis788] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The Global Enteric Multicenter Study (GEMS) is an investigation of the burden (number of cases and incidence) of moderate-to-severe diarrhea (MSD) in children <60 months of age at 7 sites in sub-Saharan Africa and South Asia. The population attributable fraction for a putative pathogen, either unadjusted or adjusted for other pathogens, is estimated using the proportion of MSD cases from whom the pathogen was isolated and the odds ratio for MSD and the pathogen from conditional logistic regression modeling. The adjusted attributable fraction, proportion of MSD cases taken to a sentinel health center (SHC), number of cases presenting to an SHC, and the site's population are used to estimate the annual number of MSD cases and MSD incidence rate attributable to a pathogen or group of pathogens. Associations with death and nutritional outcomes, ascertained at follow-up visits to case and control households, are evaluated both in MSD cases and in the population.
Collapse
Affiliation(s)
- William C Blackwelder
- Center for Vaccine Development, University of Maryland School of Medicine, 685 W Baltimore St, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Mitra S, Chakrabarti MK, Koley H. Multi-serotype outer membrane vesicles of Shigellae confer passive protection to the neonatal mice against shigellosis. Vaccine 2013; 31:3163-73. [DOI: 10.1016/j.vaccine.2013.05.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 03/14/2013] [Accepted: 05/01/2013] [Indexed: 12/27/2022]
|
28
|
Chassagne P, Fontana C, Guerreiro C, Gauthier C, Phalipon A, Widmalm G, Mulard LA. Structural Studies of theO-Acetyl-Containing O-Antigen from aShigella flexneriSerotype 6 Strain and Synthesis of Oligosaccharide Fragments Thereof. European J Org Chem 2013. [DOI: 10.1002/ejoc.201300180] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
29
|
Lipopolysaccharide modifications of a cholera vaccine candidate based on outer membrane vesicles reduce endotoxicity and reveal the major protective antigen. Infect Immun 2013; 81:2379-93. [PMID: 23630951 DOI: 10.1128/iai.01382-12] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The causative agent of the life-threatening gastrointestinal infectious disease cholera is the Gram-negative, facultative human pathogen Vibrio cholerae. We recently started to investigate the potential of outer membrane vesicles (OMVs) derived from V. cholerae as an alternative approach for a vaccine candidate against cholera and successfully demonstrated the induction of a long-lasting, high-titer, protective immune response upon immunization with OMVs using the mouse model. In this study, we present immunization data using lipopolysaccharide (LPS)-modified OMVs derived from V. cholerae, which allowed us to improve and identify the major protective antigen of the vaccine candidate. Our results indicate that reduction of endotoxicity can be achieved without diminishing the immunogenic potential of the vaccine candidate by genetic modification of lipid A. Although the protective potential of anti-LPS antibodies has been suggested many times, this is the first comprehensive study that uses defined LPS mutants to characterize the LPS-directed immune response of a cholera vaccine candidate in more detail. Our results pinpoint the O antigen to be the essential immunogenic structure and provide a protective mechanism based on inhibition of motility, which prevents a successful colonization. In a detailed analysis using defined antisera, we can demonstrate that only anti-O antigen antibodies, but not antibodies directed against the major flagellar subunit FlaA or the most abundant outer membrane protein, OmpU, are capable of effectively blocking the motility by binding to the sheathed flagellum and provide protection in a passive immunization assay.
Collapse
|
30
|
Yang Y, Wan C, Xu H, Aguilar ZP, Tan Q, Xu F, Lai W, Xiong Y, Wei H. Identification of an outer membrane protein of Salmonella enterica serovar Typhimurium as a potential vaccine candidate for Salmonellosis in mice. Microbes Infect 2013; 15:388-98. [PMID: 23485513 DOI: 10.1016/j.micinf.2013.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Revised: 02/10/2013] [Accepted: 02/18/2013] [Indexed: 12/27/2022]
Abstract
We report our investigation of the functions of PagN in Salmonella pathogenesis and its potential as a vaccine candidate. Further investigation conducted in this study indicates that the outer membrane protein PagN is important for Salmonella adhesion/invasion of epithelial cells as well as bacterial virulence. When pagN was deleted from Salmonella enterica serovar Typhimurium (S. Typhimurium), the adhesion and invasion of HT-29 epithelial cells was significantly decreased compared with the wild type strain. Mice infected with the pagN mutant strain exhibited less pathological signs in the intestine and survived longer than the wild-type-infected mice. PagN is widely distributed and conserved among clinical isolates of different Salmonella serovars, making PagN a potential vaccine candidate for Salmonella infection. To elucidate the potential of PagN as a vaccine, we expressed and purified recombinant PagN (rPagN). When rPagN was tested in mice, it provided significant protection against Salmonella infection in vivo. In vitro, anti-PagN serum enhanced clearance of Salmonella, indicating a contribution of PagN-specific antibodies to the killing process. This correlates well with the observed protection of mice immunized with rPagN. Our preliminary results indicate more functions of PagN in S. Typhimurium virulence as well as its potential as a protective vaccine.
Collapse
Affiliation(s)
- Youjun Yang
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW This review summarizes the recent advances in vaccination against Salmonella enterica serovar Typhi and highlights the data supporting the development of next generation vaccines to address paratyphoid fever and invasive nontyphoidal Salmonella (iNTS) disease. RECENT FINDINGS There has been increasing awareness of the disease burden caused by S. Typhi particularly in Africa and greater recognition of S. Paratyphi A's contribution to enteric fever episodes throughout Asia. Groups have been working to improve the existing typhoid vaccines and provide comprehensive data on the feasibility of their implementation in endemic settings. These data have resulted in modifications to the recommendations for typhoid vaccination in traveller markets and endemic settings, and has also led to the development of S. Paratyphi A vaccine components that can be combined with existing typhoid vaccines to generate bivalent formulations against enteric fever. The epidemiology of iNTS serovars as cause of appreciable morbidity and mortality in Africa, and the need for vaccines, has also become more widely appreciated. SUMMARY Current typhoid vaccines, although moderately effective for short periods of time, cannot be used in all age groups and only target one of the clinically relevant Salmonella serovars. Greater effort must be placed on the development and implementation of improved vaccines for the disease burden resulting from Typhi, Paratyphi A or iNTS infections.
Collapse
|
32
|
Soltani M, Bouanene I, Trabelsi A, Harbi A, Hachicha M, Amri F, Boussnina S, Gueddiche MN, Sfar MT, Teleb N, Ben Ghorbel M, Ben Hamida E. [Epidemiology of rotavirus gastroenteritis among children under 5 years of age in Tunisia - results of sentinel hospital surveillance 2009 to 2011]. Rev Epidemiol Sante Publique 2012; 60:473-80. [PMID: 23141818 DOI: 10.1016/j.respe.2012.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 03/02/2012] [Accepted: 04/05/2012] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Rotavirus is the major cause of severe acute gastroenteritis among young children. The objectives of this study were to assess the epidemiology, clinical and virological features of community-acquired rotavirus acute gastroenteritis, in children under 5 years of age, hospitalized in Tunisia. METHODS A multicenter prospective observational study was conducted from April 2009 to March 2011, in 11 sentinel pediatric departments. Clinical data and stool samples were collected for all children under 5 years, admitted for acute gastroenteritis. Rotavirus was detected by Elisa immunoassay test and genotyped for G and P by semi-nested multiplex RT-PCR. RESULT A total of 621 children were enrolled in this study. Rotavirus was detected in 30.3% of cases (95% CI [26.7-33.9]). The estimated incidence rate of rotavirus acute gastroenteritis was 11 cases/100,000 child-years (95% CI [9.43-12.57]). This infection affected predominantly children aged under 24 months, and occurred mainly in winter (55.3%). Vomiting, fever and dehydration were observed in 79.6%, 69.5% and 57% respectively. Genotype analysis identified four G types (G1, G2, G3 and G4) and 4 P types (P[4], P[6], P[8] and P[9]). The most common G/P combination was G3P[8] (24.4%), followed by G4P[8] (13.3%) and G1P[8] (6.5%). CONCLUSION These results highlight the frequency and potential severity of rotavirus acute gastroenteritis in pediatric hospital settings. The present study could provide a sufficient database to make a decision related to the introduction of rotavirus vaccine in Tunisian national immunization program.
Collapse
Affiliation(s)
- M Soltani
- Service de médecine préventive et d'épidémiologie, CHU Fattouma Bourguiba, 5000 Monastir, Tunisie.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Periaswamy B, Maier L, Vishwakarma V, Slack E, Kremer M, Andrews-Polymenis HL, McClelland M, Grant AJ, Suar M, Hardt WD. Live attenuated S. Typhimurium vaccine with improved safety in immuno-compromised mice. PLoS One 2012; 7:e45433. [PMID: 23029007 PMCID: PMC3454430 DOI: 10.1371/journal.pone.0045433] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 08/17/2012] [Indexed: 12/23/2022] Open
Abstract
Live attenuated vaccines are of great value for preventing infectious diseases. They represent a delicate compromise between sufficient colonization-mediated adaptive immunity and minimizing the risk for infection by the vaccine strain itself. Immune defects can predispose to vaccine strain infections. It has remained unclear whether vaccine safety could be improved via mutations attenuating a vaccine in immune-deficient individuals without compromising the vaccine's performance in the normal host. We have addressed this hypothesis using a mouse model for Salmonella diarrhea and a live attenuated Salmonella Typhimurium strain (ssaV). Vaccination with this strain elicited protective immunity in wild type mice, but a fatal systemic infection in immune-deficient cybb−/−nos2−/− animals lacking NADPH oxidase and inducible NO synthase. In cybb−/−nos2−/− mice, we analyzed the attenuation of 35 ssaV strains carrying one additional mutation each. One strain, Z234 (ssaV SL1344_3093), was >1000-fold attenuated in cybb−/−nos2−/− mice and ≈100 fold attenuated in tnfr1−/− animals. However, in wt mice, Z234 was as efficient as ssaV with respect to host colonization and the elicitation of a protective, O-antigen specific mucosal secretory IgA (sIgA) response. These data suggest that it is possible to engineer live attenuated vaccines which are specifically attenuated in immuno-compromised hosts. This might help to improve vaccine safety.
Collapse
Affiliation(s)
| | - Lisa Maier
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | | | - Emma Slack
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | | | | | - Michael McClelland
- The Vaccine Research Institute of San Diego, San Diego, California, United States of America
| | - Andrew J. Grant
- Department of Veterinary Medicine and Cambridge Infectious Diseases Consortium, University of Cambridge, Cambridge, United Kingdom
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | | |
Collapse
|
34
|
Insights from natural infection-derived immunity to cholera instruct vaccine efforts. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:1707-11. [PMID: 22993412 DOI: 10.1128/cvi.00543-12] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
35
|
Lee SJ, Liang L, Juarez S, Nanton MR, Gondwe EN, Msefula CL, Kayala MA, Necchi F, Heath JN, Hart P, Tsolis RM, Heyderman RS, MacLennan CA, Felgner PL, Davies DH, McSorley SJ. Identification of a common immune signature in murine and human systemic Salmonellosis. Proc Natl Acad Sci U S A 2012; 109:4998-5003. [PMID: 22331879 PMCID: PMC3324033 DOI: 10.1073/pnas.1111413109] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Despite the importance of Salmonella infections in human and animal health, the target antigens of Salmonella-specific immunity remain poorly defined. We have previously shown evidence for antibody-mediating protection against invasive Salmonellosis in mice and African children. To generate an overview of antibody targeting in systemic Salmonellosis, a Salmonella proteomic array containing over 2,700 proteins was constructed and probed with immune sera from Salmonella-infected mice and humans. Analysis of multiple inbred mouse strains identified 117 antigens recognized by systemic antibody responses in murine Salmonellosis. Importantly, many of these antigens were independently identified as target antigens using sera from Malawian children with Salmonella bacteremia, validating the study of the murine model. Furthermore, vaccination with SseB, the most prominent antigenic target in Malawian children, provided mice with significant protection against Salmonella infection. Together, these data uncover an overlapping immune signature of disseminated Salmonellosis in mice and humans and provide a foundation for the generation of a protective subunit vaccine.
Collapse
Affiliation(s)
- Seung-Joo Lee
- Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, and
| | - Li Liang
- Department of Medicine, University of California, Irvine, CA 92697
| | - Silvia Juarez
- Department of Medicine, University of California, Irvine, CA 92697
| | - Minelva R. Nanton
- Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, and
| | - Esther N. Gondwe
- Malawi-Liverpool-Wellcome Trust Clinical Research Program, College of Medicine, University of Malawi, PO Box 30096, Chichiri, Blantyre 3, Malawi
| | - Chisomo L. Msefula
- Malawi-Liverpool-Wellcome Trust Clinical Research Program, College of Medicine, University of Malawi, PO Box 30096, Chichiri, Blantyre 3, Malawi
| | - Matthew A. Kayala
- Institute for Genomics and Bioinformatics, University of California, Irvine, CA 92067
| | - Francesca Necchi
- Novartis Vaccines Institute for Global Health, 53100 Siena, Italy
- Medical Research Council Centre for Immune Regulation, School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; and
| | - Jennifer N. Heath
- Medical Research Council Centre for Immune Regulation, School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; and
| | - Peter Hart
- Novartis Vaccines Institute for Global Health, 53100 Siena, Italy
- Medical Research Council Centre for Immune Regulation, School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; and
| | - Renée M. Tsolis
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA 95616
| | - Robert S. Heyderman
- Malawi-Liverpool-Wellcome Trust Clinical Research Program, College of Medicine, University of Malawi, PO Box 30096, Chichiri, Blantyre 3, Malawi
- Liverpool School of Tropical Medicine, University of Liverpool, Liverpool L3 5QA, United Kingdom
| | - Calman A. MacLennan
- Malawi-Liverpool-Wellcome Trust Clinical Research Program, College of Medicine, University of Malawi, PO Box 30096, Chichiri, Blantyre 3, Malawi
- Novartis Vaccines Institute for Global Health, 53100 Siena, Italy
- Medical Research Council Centre for Immune Regulation, School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; and
| | | | - D. Huw Davies
- Department of Medicine, University of California, Irvine, CA 92697
| | - Stephen J. McSorley
- Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, and
| |
Collapse
|
36
|
López-Gigosos RM, Plaza E, Díez-Díaz RM, Calvo MJ. Vaccination strategies to combat an infectious globe: oral cholera vaccines. J Glob Infect Dis 2011; 3:56-62. [PMID: 21572610 PMCID: PMC3068580 DOI: 10.4103/0974-777x.77297] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cholera is a substantial health burden in many countries in Africa and Asia, where it is endemic. It is as well responsible for ongoing epidemics in sub-Saharan Africa which are becoming greater in terms of frequency, extension, and duration. Given the availability of two oral cholera vaccines and the new data on their efficacy, field effectiveness, feasibility, and acceptance in cholera-affected populations and in travelers, these vaccines should be used in endemic areas, in travelers for these areas and should be considered in areas at risk for outbreaks. The two vaccines currently available in worldwide are: (1) The killed oral vaccine (Dukoral, licensed by SBL–Sweden to Crucell–Holland) is recommended since 1999 by WHO and consists of a mixture of four preparations of heat or formalin killed whole cell Vibrio cholera O1 (Inaba and Ogaba serotypes, and classical and El Tor biotypes) that are then added with purified recombinant cholera toxin (CT) B subunit. Because CT cross-reacts with Escherichia coli LT the vaccine also provides short-term protection against ETEC (enterotoxigenic E. coli) which is of added benefit for travelers. It is available in more than 60 countries. (2) A bivalent O1 and O139 whole cell oral vaccine without CT B subunit (Shanchol) has been lately developed in Vietnam (licensed by VaBiotech–Viet Nam to Shantha Biotechnics–India. It is available in India and Indonesia. A structured search of papers in PubMed and reports on cholera vaccines by WHO and CDC, as well as critical reading and synthesis of the information was accomplished. Inclusion criteria were defined according to reports quality and relevance.
Collapse
Affiliation(s)
- Rosa M López-Gigosos
- International Vaccination Centre of Malaga, Ministry of Health, Subdelegation in Malaga, Paseo Marítimo Pablo Ruiz, Picasso Malaga
| | | | | | | |
Collapse
|
37
|
Development of protective immunity to Salmonella, a mucosal pathogen with a systemic agenda. Mucosal Immunol 2011; 4:371-82. [PMID: 21307847 PMCID: PMC4084725 DOI: 10.1038/mi.2011.2] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Salmonella infections can cause a range of intestinal and systemic diseases in human and animal hosts. Although some Salmonella serovars initiate a localized intestinal inflammatory response, others use the intestine as a portal of entry to initiate a systemic infection. Considerable progress has been made in understanding bacterial invasion and dissemination strategies, as well as the nature of the Salmonella-specific immune response to oral infection. Innate and adaptive immunity are rapidly initiated after oral infection, but these effector responses can also be hindered by bacterial evasion strategies. Furthermore, although Salmonella resides within intramacrophage phagosomes, recent studies have highlighted a surprising collaboration of CD4 Th1, Th17, and B-cell responses in mediating resistance to Salmonella infection.
Collapse
|
38
|
Desai SN, Sahastrabuddhe S, Ochiai RL, Wierzba TF. Enteric vaccines for resource-limited countries: current status and future prospects. Pediatr Ann 2011; 40:351-7. [PMID: 21736257 DOI: 10.3928/00904481-20110615-07] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Sachin N Desai
- Intrenational Vaccine Institute, SNU Research Park, Seoul, Korea.
| | | | | | | |
Collapse
|
39
|
Bisi-Johnson MA, Obi CL, Vasaikar SD, Baba KA, Hattori T. Molecular basis of virulence in clinical isolates of Escherichia coli and Salmonella species from a tertiary hospital in the Eastern Cape, South Africa. Gut Pathog 2011; 3:9. [PMID: 21663681 PMCID: PMC3125331 DOI: 10.1186/1757-4749-3-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 06/10/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Apart from localized gastrointestinal infections, Escherichia coli and Salmonella species are major causes of systemic disease in both humans and animals. Salmonella spp. cause invasive infections such as enteric fever, septicemia, osteomyelitis and meningitis while certain types of E. coli can cause systemic infections, includingpyelonephritis, meningitis and septicemia. These characteristic requires the involvement of a myriad of virulence factors. METHODS This study investigated the virulence factors of Escherichia coli and Salmonella species in clinical specimens from patients with diarrhoea presenting to health care centres in Oliver R. Tambo District Municipality, Eastern Cape Province, Republic of South Africa. Microbiology analysis involved the use of cultural and molecular techniques. RESULTS Out of a total of 315 samples screened, Salmonella isolates were obtained in 119 (37.8%) of cases and these comprised: S. choleraesuis (6%), S. enteritidis (4%), S. eppendorf (1%), S. hadar (1%), S. isangi (8%), S. panama (1%), S. typhi (52%), S. typhimurium (25%) and untyped Salmonella spp. (2%). Among the Salmonella species 87 (73.1%) were invasive. Using molecular diagnostic methods, diarrheagenic E. coli were detected in 90 cases (28.6%): the greater proportion of this were enteroaggregative E. coli (EAEC) 37 (41.1%), enteropathogenic E. coli (EPEC) 21 (23.3%) and enterohemorrhagic E. coli (EHEC) 21 (23.3%). The predominant virulence gene among the diarrheagenic E. coli was EAEC heat-stable enterotoxin astA genes while the virulence genes identified in the Salmonella strains were 15 (12.6%) flic and 105 (88.2%) inv genes. The amino acid identity of the representative genes showed 95-100% similarity to corresponding blast searched sequence. CONCLUSIONS This study showed the diversity of virulence gene expression in two major enteric pathogens. S. typhi and enteroaggregative E. coli were the predominant enteropathogens in our study area with an indication that EAEC is endemic within our study population. It was observed among other things that some diarrheagenic E. coli isolated from apparently asymptomatic subjects expressed some virulence genes at frequency as high as seen in diarrheagenic cases. This study underlines the importance of understanding the virulence composition and diversity of pathogens for enhanced clinico-epidemiological monitoring and health care delivery.
Collapse
Affiliation(s)
- Mary A Bisi-Johnson
- Department of Medical Microbiology, Walter Sisulu University, Mthatha 5117, South Africa.
| | | | | | | | | |
Collapse
|
40
|
Nunes M, Penna F, Franco R, Mendes E, Magalhães P. Enterotoxigenic Escherichia coli in children with acute diarrhoea and controls in Teresina/PI, Brazil: distribution of enterotoxin and colonization factor genes. J Appl Microbiol 2011; 111:224-32. [DOI: 10.1111/j.1365-2672.2011.05031.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
41
|
Abstract
Bioterrorism is the deliberate release of biological toxins, pathogenic viruses, bacteria, parasites, or other infectious agents into the public sphere with the objective of causing panic, illness, and/or death on a local, regional, or possibly national scale. The list of potential biological agents compiled by the Centers for Disease Control and Prevention is long and diverse. However, a trait common to virtually all the potential bioterrorism agents is the fact that they are likely to be disseminated by either aerosol or in food/water supplies with the intention of targeting the mucosal surfaces of the respiratory or gastrointestinal tracts, respectively. In some instances, inhalation or ingestion would mimic the natural route by which humans are exposed to these agents. In other instances, (e.g., the inhalation of a toxin is normally associated with food borne illness), it would represent an unnatural route of exposure. For most potential bioterrorism agents, the respiratory or gastrointestinal mucosa may simply serve as a route of entry by which they gain access to the systemic compartment where intoxication/replication occurs. For others, however, the respiratory or gastrointestinal mucosa is the primary tissue associated with pathogenesis, and therefore, the tissue for which countermeasures must be developed.
Collapse
Affiliation(s)
- Pamela A. Kozlowski
- , Louisiana Gene Therapy Research Consorti, LSU Health Sciences Center, Poydras Street 1615, New Orleans, 70112 Louisiana USA
| | | | | |
Collapse
|
42
|
Rajendran P, Ajjampur SSR, Chidambaram D, Chandrabose G, Thangaraj B, Sarkar R, Samuel P, Rajan DP, Kang G. Pathotypes of diarrheagenic Escherichia coli in children attending a tertiary care hospital in South India. Diagn Microbiol Infect Dis 2011; 68:117-22. [PMID: 20846583 DOI: 10.1016/j.diagmicrobio.2010.06.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 05/29/2010] [Accepted: 06/05/2010] [Indexed: 11/19/2022]
Abstract
The prevalence of diarrheagenic Escherichia coli (DEC) in children under 5 years was studied in children with diarrhea and controls in South India. Four polymerase chain reaction (PCR) "schemes" were used to detect genes of the 6 pathotypes of DEC. In 394 children with diarrhea, 203 (52%) DEC infections were found. Among the 198 controls, 126 (63%) DEC infections were found. Enteroaggregative E. coli was the most common pathotype by multiplex PCR both in cases (58, 14.7%) and controls (47, 23.7%), followed by enteropathogenic E. coli seen in 10% cases and 8% of controls. Enterotoxigenic E. coli (ETEC), enterohemorrhagic E. coli (EHEC), enteroinvasive E. coli (EIEC), and diffusely adherent E. coli (DAEC) were found in 4.1%, 2.0%, 1.0%, and 0.5% of cases, respectively. ETEC was found in 2.5% of controls, but EHEC, EIEC, and DAEC were not detected. Overall, no single assay worked well, but by discounting genes with a pathogenicity index of less than 1, it was possible to use the PCR assays to identify DEC in 75/394 (19%) cases and 12/198 (6.1%) controls, while mixed infection could be identified in 8/394 (2%) cases and 2/198 (1%) controls.
Collapse
Affiliation(s)
- Priya Rajendran
- Department of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Snelling TL, Andrews RM, Kirkwood CD, Culvenor S, Carapetis JR. Case-Control Evaluation of the Effectiveness of the G1P[8] Human Rotavirus Vaccine during an Outbreak of Rotavirus G2P[4] Infection in Central Australia. Clin Infect Dis 2010; 52:191-9. [DOI: 10.1093/cid/ciq101] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
44
|
Kelly DF, Thorson S, Maskey M, Mahat S, Shrestha U, Hamaluba M, Williams E, Dongol S, Werno AM, Portess H, Yadav BK, Adhikari N, Guiver M, Thomas K, Murdoch DR, Pollard AJ. The burden of vaccine-preventable invasive bacterial infections and pneumonia in children admitted to hospital in urban Nepal. Int J Infect Dis 2010; 15:e17-23. [PMID: 21123100 DOI: 10.1016/j.ijid.2010.05.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 01/30/2010] [Accepted: 05/10/2010] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Protein-polysaccharide vaccines have made a significant impact on the burden of disease caused by encapsulated bacteria such as Streptococcus pneumoniae, Haemophilus influenzae, Neisseria meningitidis, and have the potential to do so for Salmonella Typhi. Nepal is one of many resource-poor nations with limited information on the epidemiology of childhood infections caused by these pathogens. METHODS Over a 21-month period, we studied children aged ≤12 years admitted to an urban hospital in Nepal with suspected bacteremia, meningitis, or pneumonia. Patan Hospital is a non-profit hospital with the second largest pediatric unit in the Kathmandu Valley. RESULTS Of 2039 children enrolled in the study, 142 (7.5%) included in the analysis had positive blood cultures. The agents of enteric fever, Salmonella Typhi and Salmonella Paratyphi, accounted for 59/142 (42%) of all bacteremias and were the most frequently cultured pathogens in children ≥1 year of age. S. pneumoniae was isolated in 16% of positive blood cultures and was the most common cause of bacteremia in children <1 year of age. Pneumonia accounted for 51% of admissions in children ≥2 months, with 44% of these children having radiographically defined primary endpoint pneumonia. S. pneumoniae was the most commonly identified pathogen in cases of pneumonia and meningitis. The S. pneumoniae serotype distribution indicated that the 10-valent and 13-valent pneumococcal conjugate vaccines would cover 44% and 47%, respectively, of all S. pneumoniae cultured from blood or cerebrospinal fluid (CSF) isolates and 62% and 67%, respectively, of isolates associated with pneumonia. H. influenzae type b was isolated infrequently from blood or CSF cultures, but is likely to be more important as a cause of pneumonia. CONCLUSIONS The data on the burden of invasive bacterial infections and pneumonia from this study suggest that vaccines in development against Salmonella Typhi and the pneumococcus have the potential to significantly improve the health of children in Nepal.
Collapse
Affiliation(s)
- Dominic F Kelly
- Department of Paediatrics, University of Oxford, Level 2, Room 02-46-03, Children's Hospital, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Mantis NJ, Forbes SJ. Secretory IgA: arresting microbial pathogens at epithelial borders. Immunol Invest 2010; 39:383-406. [PMID: 20450284 DOI: 10.3109/08820131003622635] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Secretory IgA (SIgA) is the predominant class of antibody found in intestinal secretions. Although SIgA's role in protecting the intestinal epithelium from the enteric pathogens and toxins has long been recognized, surprisingly little is known about the molecular mechanisms by which this is achieved. The present review summarizes the current understanding of how SIgA functions to prevent microbial pathogens and toxins from gaining access to the intestinal epithelium. We also discuss recent work from our laboratory examining the interaction of a particular protective monoclonal IgA with Salmonella and propose, based on this work, that SIgA has a previously unrecognized capacity to directly interfere with microbial virulence at mucosal surfaces.
Collapse
Affiliation(s)
- Nicholas J Mantis
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, University at Albany School of Public Health, Albany, New York 12208, USA.
| | | |
Collapse
|
46
|
Edwards AD, Chatterjee P, Mahbubani KT, Reis CM, Slater NK. Optimal protection of stabilised dry live bacteria from bile toxicity in oral dosage forms by bile acid adsorbent resins. Chem Eng Sci 2010. [DOI: 10.1016/j.ces.2010.05.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
47
|
Hien TT, Dung NT, Truong NT, Van NTT, Bich Chau TN, Hoang NVM, Nga TTT, Thuy CT, Minh PV, Binh NTC, Ha TTD, Toi PV, Song Diep T, Campbell JI, Stockwell E, Schultsz C, Simmons CP, Glover C, Lam W, Marques F, May JP, Upton A, Budhram R, Dougan G, Farrar J, Vinh Chau NV, Dolecek C. A randomised trial evaluating the safety and immunogenicity of the novel single oral dose typhoid vaccine M01ZH09 in healthy Vietnamese children. PLoS One 2010; 5:e11778. [PMID: 20668668 PMCID: PMC2909895 DOI: 10.1371/journal.pone.0011778] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 06/15/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The emergence of drug resistant typhoid fever is a major public health problem, especially in Asia. An oral single dose typhoid vaccine would have major advantages. M01ZH09 is a live oral single dose candidate typhoid vaccine containing Salmonella enterica serovar Typhi (Ty2 aroC(-)ssaV(-)) ZH9 with two independently attenuating deletions. Studies in healthy adults demonstrated immunogenicity and an acceptable safety profile. OBJECTIVES We conducted a randomised placebo controlled, single-blind trial to evaluate the safety and immunogenicity of M01ZH09 in healthy Vietnamese children aged 5 to 14 years. METHODS Subjects were randomly assigned to receive either a nominal dose of 5x10(9) CFU of M01ZH09 or placebo and were followed up for 28 days. The primary safety outcome was the proportion of subjects with any adverse event attributed to M01ZH09. The primary immunogenicity endpoint was the proportion of subjects who showed a positive immune response to M01ZH09 in the Salmonella Typhi lipopolysaccharide (LPS) specific serum IgA and IgG ELISA. PRINCIPAL FINDINGS One hundred and fifty-one children were enrolled, 101 subjects received M01ZH09 and 50 subjects received placebo. An intention to treat analysis was conducted. There were no serious adverse events and no bacteraemias. In the M01ZH09 group, 26 (26%; 95% CI, 18-5%) of 101 subjects experienced adverse events compared to 11 (22%; 95% CI, 12-36%) of 50 subjects in the placebo group (odds ratio (OR) [95%CI] = 1.23 [0.550-2.747]; p = 0.691). Faecal shedding of S. Typhi (Ty2 aroC(-)ssaV(-)) ZH9 was detected in 51 (51%; 95% CI, 41-61%) of 100 M01ZH09 subjects. No shedding was detected beyond day 3. A positive immune response, defined as 70% increase (1.7 fold change) in LPS specific serum IgG (day 14 or 28) and/or 50% increase (1.5 fold change) in LPS specific serum IgA (day 7 or 14) from baseline was detected in 98 (97%; 95% CI, 92-99%) of 101 M01ZH09 recipients and 8 (16%; 95% CI, 7-29%) of 50 placebo recipients. Twenty-eight (100%; 95% CI, 88-100%) of 28 vaccine recipients who were evaluated in the LPS specific IgA ELISPOT assay showed a positive response compared to none of the 14 placebo recipients tested. CONCLUSIONS This was the first phase II trial of a novel oral candidate typhoid vaccine in children in an endemic country. M01ZH09 had an appropriate safety profile and was immunogenic in children. TRIAL REGISTRATION Controlled-trials.com ISRCTN91111837.
Collapse
Affiliation(s)
- Tran Tinh Hien
- The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Nguyen Thi Dung
- The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | | | - Ninh Thi Thanh Van
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Tran Nguyen Bich Chau
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Nguyen Van Minh Hoang
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Tran Thi Thu Nga
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Cao Thu Thuy
- The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Pham Van Minh
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Nguyen Thi Cam Binh
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Tran Thi Diem Ha
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Pham Van Toi
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - To Song Diep
- The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - James I. Campbell
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine, University of Oxford, Oxford, United Kingdom
| | - Elaine Stockwell
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine, University of Oxford, Oxford, United Kingdom
| | - Constance Schultsz
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine, University of Oxford, Oxford, United Kingdom
- Academic Medical Center, Center for Poverty-related Communicable Diseases, Amsterdam, The Netherlands
| | - Cameron P. Simmons
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Winnie Lam
- Emergent Product Development UK Ltd., Wokingham, United Kingdom
| | - Filipe Marques
- Emergent Product Development UK Ltd., Wokingham, United Kingdom
| | - James P. May
- Emergent Product Development UK Ltd., Wokingham, United Kingdom
| | - Anthony Upton
- Emergent Product Development UK Ltd., Wokingham, United Kingdom
| | - Ronald Budhram
- Emergent Product Development UK Ltd., Wokingham, United Kingdom
| | - Gordon Dougan
- The Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Jeremy Farrar
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine, University of Oxford, Oxford, United Kingdom
| | - Nguyen Van Vinh Chau
- The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Christiane Dolecek
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine, University of Oxford, Oxford, United Kingdom
- The London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
48
|
Genotypic and phenotypic characterization of enterotoxigenic Escherichia coli strains isolated from Peruvian children. J Clin Microbiol 2010; 48:3198-203. [PMID: 20631096 DOI: 10.1128/jcm.00644-10] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major cause of childhood diarrhea. The present study sought to determine the prevalence and distribution of toxin types, colonization factors (CFs), and antimicrobial susceptibility of ETEC strains isolated from Peruvian children. We analyzed ETEC strains isolated from Peruvian children between 2 and 24 months of age in a passive surveillance study. Five E. coli colonies per patient were studied by multiplex real-time PCR to identify ETEC virulence factors. ETEC-associated toxins were confirmed using a GM1-based enzyme-linked immunosorbent assay. Confirmed strains were tested for CFs by dot blot assay using 21 monoclonal antibodies. We analyzed 1,129 samples from children with diarrhea and 744 control children and found ETEC in 5.3% and 4.3%, respectively. ETEC was more frequently isolated from children >12 months of age than from children <12 months of age (P < 0.001). Fifty-two percent of ETEC isolates from children with diarrhea and 72% of isolates from controls were heat-labile enterotoxin (LT) positive and heat-stable enterotoxin (ST) negative; 25% and 19%, respectively, were LT negative and ST positive; and 23% and 9%, respectively, were LT positive and ST positive. CFs were identified in 64% of diarrheal samples and 37% of control samples (P < 0.05). The most common CFs were CS6 (14% and 7%, respectively), CS12 (12% and 4%, respectively), and CS1 (9% and 4%, respectively). ST-producing ETEC strains caused more severe diarrhea than non-ST-producing ETEC strains. The strains were most frequently resistant to ampicillin (71%) and co-trimoxazole (61%). ETEC was thus found to be more prevalent in older infants. LT was the most common toxin type; 64% of strains had an identified CF. These data are relevant in estimating the burden of disease due to ETEC and the potential coverage of children in Peru by investigational vaccines.
Collapse
|
49
|
de Palma O, Cruz L, Ramos H, de Baires A, Villatoro N, Pastor D, de Oliveira LH, Kerin T, Bowen M, Gentsch J, Esposito DH, Parashar U, Tate J, Patel M. Effectiveness of rotavirus vaccination against childhood diarrhoea in El Salvador: case-control study. BMJ 2010; 340:c2825. [PMID: 20551120 PMCID: PMC2886195 DOI: 10.1136/bmj.c2825] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To evaluate the effectiveness of a monovalent rotavirus vaccine against severe rotavirus disease and to assess its impact on diarrhoea in children aged less than 2 years after national introduction in El Salvador, a low-middle income country in Central America. DESIGN Matched case-control study. SETTING Seven hospitals in cities across El Salvador, January 2007 to June 2009. PARTICIPANTS 323 children aged less than 2 years admitted with laboratory confirmed rotavirus diarrhoea and 969 healthy controls matched for age and neighbourhood. MAIN OUTCOME MEASURE Effectiveness of rotavirus vaccination ((1-adjusted odds ratio of vaccination)x100) against rotavirus diarrhoea requiring hospital admission. RESULTS Cases and controls were similar for breast feeding, premature birth, maternal education, and socioeconomic variables. G1P[8] strains were identified in 92% of rotavirus cases. Effectiveness of two doses of vaccination against diarrhoea requiring hospital admission was 76% (95% confidence interval 64% to 84%). Protection was significantly lower (P=0.046) among children aged 12 months or more (59%, 27% to 77%) compared with children aged 6-11 months (83%, 68% to 91%). One dose of vaccine was 51% (26% to 67%) effective. At the sentinel hospitals, all admissions for diarrhoea among children under 5 declined by 40% in 2008 and by 51% in 2009 from the prevaccine year 2006. CONCLUSIONS A monovalent rotavirus vaccine was highly effective against admissions for rotavirus diarrhoea in children aged less than 2 years in El Salvador and substantially reduced the number of such admissions in this low-middle income setting. The impact on disease epidemiology after vaccination, particularly among older children, warrants future attention.
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW To summarize recent advances that contribute to the development of new vaccines against gastrointestinal infections. RECENT FINDINGS The main themes of this review include epidemiology assessing disease burden of enteric pathogens, increasing antibiotic resistance, positive effect of existing enteric vaccines in developed and developing countries, and recommendations of advisory bodies; antigen discovery and preclinical testing, including live vector systems expressing heterologous antigens, conjugation of antigen to carrier proteins, and plant-based expression systems; as well as clinical studies highlighting recently published enteric vaccine candidates, studies assessing immune correlates of protection, as well as a trend for technology transfer to developing countries where enteric vaccines can be produced less expensively. SUMMARY It is an exciting time for the development of novel vaccines against gastrointestinal infections. Better understanding of disease burden, interest from funding sources, identification of novel antigens, better understanding of protective immune responses, and steady progress in the conduct of clinical trials make the development of new enteric vaccines a goal that is attainable in the near future.
Collapse
|