1
|
Frem JA, Russell A, Fitzpatrick C, Williams D, Richardson D. Gastrointestinal Escherichia coli in men who have sex with men: A systematic review. Int J STD AIDS 2025; 36:176-184. [PMID: 39648861 DOI: 10.1177/09564624241306847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
OBJECTIVE This systematic review aimed to explore any demographic, biological and behavioural characteristics of men who have sex with men (MSM) with diarrhoeagenic E.coli. DESIGN/METHODS We searched MEDLINE, EMBASE, and CINAHL for manuscripts published to March 2024. One author screened manuscript abstracts; two authors independently conducted a full text review. We only included primary data on gastrointestinal E.coli in MSM. Risk of bias was assessed independently by two authors using the Joanna Briggs Institute tools. This review was registered on PROSPERO(CRD42023455321). RESULTS Eleven manuscripts (cross-sectional studies (n = 8), case-series (n = 1), case-control study (n = 1), longitudinal study (n = 1)) from Europe (n = 7) Australia (n = 2), USA (n = 2) including 983 MSM with gastrointestinal E.coli published between 2014-2023 were included in this review. Demographic factors (living with HIV, using HIV-PrEP, using dating apps and working as airline crew, group sex, non-regular (casual) sexual partners); behavioural factors (non-regular sexual partners, non-condom use, oro-anal sex, penile-anal sex, use of sex toys, insertive and receptive fisting, scat play); and infection factors (co-infection with Chlamydia trachomatis including LGV, Neisseria gonorrhoeae, Treponema pallidum, hepatitis C, other enteric pathogens [Shigella spp. Giardia duodenalis, Entamoeba histolytica, hepatitis A and intestinal spirochaetosis]) were observed in MSM with E. coli. Antimicrobial resistance (extended spectrum beta-lactamase and quinolone resistance) was described in MSM with E.coli. CONCLUSION We have highlighted demographic, behavioral and infection factors observed in MSM with E.coli suggesting sexual transmissibility. These data provide insight for future clinical guidelines, public health control strategies and research.
Collapse
Affiliation(s)
- Jim Abi Frem
- Sexual health & HIV, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
| | - Annie Russell
- Sexual health & HIV, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
| | - Colin Fitzpatrick
- Sexual health & HIV, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
| | - Deborah Williams
- Sexual health & HIV, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
| | - Daniel Richardson
- Sexual health & HIV, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
- Brighton & Sussex Medical School, Brighton UK
| |
Collapse
|
2
|
Maciel M, Scott JC, Baudier RL, Clements JD, Laird RM, Gutiérrez RL, Porter CK, Norton EB. Protective antibodies against enterotoxigenic Escherichia coli are generated from heat-labile toxoid vaccination and exhibit subject- and vaccine-specific diversity. Med Microbiol Immunol 2025; 214:10. [PMID: 39934422 PMCID: PMC11814043 DOI: 10.1007/s00430-025-00817-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/22/2025] [Indexed: 02/13/2025]
Abstract
Heat-labile toxin (LT) from enterotoxigenic Escherichia coli (ETEC) is an important pathogenic protein. Anti-LT antibodies (Abs) induced by vaccination can neutralize the toxin and potentially prevent diarrheal secretion from ~ 60% of ETEC strains expressing LT. However, only superficial investigation of the anti-toxin response is usually conducted in clinical trials. Here, we utilized human serum samples from two clinical trials performed to assess safety, immunogenicity and protection in a controlled human infection model with a LT + ST + CFA/I + H10407 ETEC strain. These Phase 1 and Phase 2b clinical trials explored a prototype ETEC adhesin (CfaE) and a chimeric adhesin-toxoid protein (dscCfaE-CTA2/LTB5) delivered intradermally or transcutaneously with a mutated form of LT (mLT) as an adjuvant. Serum samples were tested for antigen-specific IgG or IgA Abs by immunoblot, enzyme-linked immunosorbent assay (ELISA), or functional neutralizing Abs using LT holotoxin, LTA or LTB subunits. Abs to both LT subunits were present, but the response to each was altered by vaccine formulation, dose, and delivery routes as well as subject. The anti-LT IgG response correlated best to neutralizing antibodies and protection from H10407 controlled challenge when compared to other measures including serum IgA or anti-fimbriae (CfaE) Abs. In addition, our results helped to explain cohort attack rate differences in naïve unvaccinated participants and we found higher anti-LTA IgG post-challenge significantly related to ETEC severity score. Thus, strategies generating and measuring immunity to the complete AB5 structure of LT and subunits are better determinant of assessing protective immunity against LT + or LT + ST + ETEC diarrheal secretion in humans.
Collapse
Affiliation(s)
- Milton Maciel
- Operationally Relevant Infections Department, Naval Medical Research Command, Silver Spring, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Jordan C Scott
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Robin L Baudier
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
- Biostatistics and Design Program, Oregon Health and Sciences University, Portland, USA
| | - John D Clements
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Renee M Laird
- Operationally Relevant Infections Department, Naval Medical Research Command, Silver Spring, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
- Diarrheal Disease Research Branch, Walter Reed Army Institute of Research, Silver Spring, USA
| | - Ramiro L Gutiérrez
- Operationally Relevant Infections Department, Naval Medical Research Command, Silver Spring, MD, USA
- State University of New York Upstate Medical University, Syracuse, USA
| | - Chad K Porter
- Translational and Clinical Research Department, Naval Medical Research Command, Silver Spring, MD, USA
| | - Elizabeth B Norton
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
3
|
Salvador-Erro J, Pastor Y, Gamazo C. Targeting Enterotoxins: Advancing Vaccine Development for Enterotoxigenic Escherichia coli ETEC. Toxins (Basel) 2025; 17:71. [PMID: 39998088 PMCID: PMC11860656 DOI: 10.3390/toxins17020071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major cause of diarrheal disease worldwide, particularly in children in low- and middle-income countries. Its ability to rapidly colonize the intestinal tract through diverse colonization factors and toxins underpins its significant public health impact. Despite extensive research and several vaccine candidates reaching clinical trials, no licensed vaccine exists for ETEC. This review explores the temporal and spatial coordination of ETEC virulence factors, focusing on the interplay between adherence mechanisms and toxin production as critical targets for therapeutic intervention. Advancements in molecular biology and host-pathogen interaction studies have uncovered species-specific variations and cross-reactivity between human and animal strains. In particular, the heat-labile (LT) and heat-stable (ST) toxins have provided crucial insights into molecular mechanisms and intestinal disruption. Additional exotoxins, such as EAST-1 and hemolysins, further highlight the multifactorial nature of ETEC pathogenicity. Innovative vaccine strategies, including multiepitope fusion antigens (MEFAs), mRNA-based approaches, and glycoconjugates, aim to enhance broad-spectrum immunity. Novel delivery methods, like intradermal immunization, show promise in eliciting robust immune responses. Successful vaccination against ETEC will offer an effective and affordable solution with the potential to greatly reduce mortality and prevent stunting, representing a highly impactful and cost-efficient solution to a critical global health challenge.
Collapse
Affiliation(s)
| | | | - Carlos Gamazo
- Department of Microbiology and Parasitology, Navarra Medical Research Institute (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (J.S.-E.); (Y.P.)
| |
Collapse
|
4
|
Gutiérrez RL, Porter CK, Harro C, Talaat K, Riddle MS, DeNearing B, Brubaker J, Maciel M, Laird RM, Poole S, Chakraborty S, Maier N, Sack DA, Savarino SJ. Efficacy Evaluation of an Intradermally Delivered Enterotoxigenic Escherichia coli CF Antigen I Fimbrial Tip Adhesin Vaccine Coadministered with Heat-Labile Enterotoxin with LT(R192G) against Experimental Challenge with Enterotoxigenic E. coli H10407 in Healthy Adult Volunteers. Microorganisms 2024; 12:288. [PMID: 38399692 PMCID: PMC10892241 DOI: 10.3390/microorganisms12020288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/16/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Enterotoxigenic E. coli (ETEC) is a principal cause of diarrhea in travelers, deployed military personnel, and children living in low to middle-income countries. ETEC expresses a variety of virulence factors including colonization factors (CF) that facilitate adherence to the intestinal mucosa. We assessed the protective efficacy of a tip-localized subunit of CF antigen I (CFA/I), CfaE, delivered intradermally with the mutant E. coli heat-labile enterotoxin, LTR192G, in a controlled human infection model (CHIM). METHODS Three cohorts of healthy adult subjects were enrolled and given three doses of 25 μg CfaE + 100 ng LTR192G vaccine intradermally at 3-week intervals. Approximately 28 days after the last vaccination, vaccinated and unvaccinated subjects were admitted as inpatients and challenged with approximately 2 × 107 cfu of CFA/I+ ETEC strain H10407 following an overnight fast. Subjects were assessed for moderate-to-severe diarrhea for 5 days post-challenge. RESULTS A total of 52 volunteers received all three vaccinations; 41 vaccinated and 43 unvaccinated subjects were challenged and assessed for moderate-to-severe diarrhea. Naïve attack rates varied from 45.5% to 64.7% across the cohorts yielding an overall efficacy estimate of 27.8% (95% confidence intervals: -7.5-51.6%). In addition to reducing moderate-severe diarrhea rates, the vaccine significantly reduced loose stool output and overall ETEC disease severity. CONCLUSIONS This is the first study to demonstrate protection against ETEC challenge after intradermal vaccination with an ETEC adhesin. Further examination of the challenge methodology is necessary to address the variability in naïve attack rate observed among the three cohorts in the present study.
Collapse
Affiliation(s)
- Ramiro L. Gutiérrez
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (M.S.R.); (R.M.L.); (S.P.); (S.J.S.)
| | - Chad K. Porter
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (M.S.R.); (R.M.L.); (S.P.); (S.J.S.)
| | - Clayton Harro
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21224, USA (K.T.); (B.D.); (D.A.S.)
| | - Kawsar Talaat
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21224, USA (K.T.); (B.D.); (D.A.S.)
| | - Mark S. Riddle
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (M.S.R.); (R.M.L.); (S.P.); (S.J.S.)
| | - Barbara DeNearing
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21224, USA (K.T.); (B.D.); (D.A.S.)
| | - Jessica Brubaker
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21224, USA (K.T.); (B.D.); (D.A.S.)
| | - Milton Maciel
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (M.S.R.); (R.M.L.); (S.P.); (S.J.S.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Renee M. Laird
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (M.S.R.); (R.M.L.); (S.P.); (S.J.S.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Steven Poole
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (M.S.R.); (R.M.L.); (S.P.); (S.J.S.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Subra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| | | | - David A. Sack
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21224, USA (K.T.); (B.D.); (D.A.S.)
| | - Stephen J. Savarino
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (M.S.R.); (R.M.L.); (S.P.); (S.J.S.)
| |
Collapse
|
5
|
Porter CK, Talaat KR, Isidean SD, Kardinaal A, Chakraborty S, Gutiérrez RL, Sack DA, Bourgeois AL. The Controlled Human Infection Model for Enterotoxigenic Escherichia coli. Curr Top Microbiol Immunol 2024; 445:189-228. [PMID: 34669040 DOI: 10.1007/82_2021_242] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The controlled human infection model (CHIM) for enterotoxigenic Escherichia coli (ETEC) has been instrumental in defining ETEC as a causative agent of acute watery diarrhea, providing insights into disease pathogenesis and resistance to illness, and enabling preliminary efficacy evaluations for numerous products including vaccines, immunoprophylactics, and drugs. Over a dozen strains have been evaluated to date, with a spectrum of clinical signs and symptoms that appear to replicate the clinical illness seen with naturally occurring ETEC. Recent advancements in the ETEC CHIM have enhanced the characterization of clinical, immunological, and microbiological outcomes. It is anticipated that omics-based technologies applied to ETEC CHIMs will continue to broaden our understanding of host-pathogen interactions and facilitate the development of primary and secondary prevention strategies.
Collapse
Affiliation(s)
- Chad K Porter
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, 20910, USA.
| | - Kawsar R Talaat
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Sandra D Isidean
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation, Bethesda, MD, 20817, USA
| | - Alwine Kardinaal
- NIZO Food Research, Ede, P.O. Box 20, 6710 BA EDE, Kernhemseweg 2, 6718 ZB EDE, The Netherlands
| | - Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Ramiro L Gutiérrez
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, 20910, USA
| | - David A Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - A Louis Bourgeois
- PATH|Center for Vaccine Innovation and Access, 455 Massachusetts Avenue NW, Suite 1000, Washington, DC, 20001, USA
| |
Collapse
|
6
|
Islam D, Ruamsap N, Imerbsin R, Khanijou P, Gonwong S, Wegner MD, McVeigh A, Poly FM, Crawford JM, Swierczewski BE, Kaminski RW, Laird RM. Bioactivity and efficacy of a hyperimmune bovine colostrum product- Travelan, against shigellosis in a non-Human primate model (Macaca mulatta). PLoS One 2023; 18:e0294021. [PMID: 38091314 PMCID: PMC10718440 DOI: 10.1371/journal.pone.0294021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 10/16/2023] [Indexed: 12/18/2023] Open
Abstract
Infectious diarrhea is a World Health Organization public health priority area due to the lack of effective vaccines and an accelerating global antimicrobial resistance crisis. New strategies are urgently needed such as immunoprophylactic for prevention of diarrheal diseases. Hyperimmune bovine colostrum (HBC) is an established and effective prophylactic for infectious diarrhea. The commercial HBC product, Travelan® (Immuron Ltd, Australia) targets multiple strains of enterotoxigenic Escherichia coli (ETEC) is highly effective in preventing diarrhea in human clinical studies. Although Travelan® targets ETEC, preliminary studies suggested cross-reactivity with other Gram-negative enteric pathogens including Shigella and Salmonella species. For this study we selected an invasive diarrheal/dysentery-causing enteric pathogen, Shigella, to evaluate the effectiveness of Travelan®, both in vitro and in vivo. Here we demonstrate broad cross-reactivity of Travelan® with all four Shigella spp. (S. flexneri, S. sonnei, S. dysenteriae and S. boydii) and important virulence factor Shigella antigens. Naïve juvenile rhesus macaques (NJRM) were randomized, 8 dosed with Travelan® and 4 with a placebo intragastrically twice daily over 6 days. All NJRM were challenged with S. flexneri 2a strain 2457T on the 4th day of treatment and monitored for diarrheal symptoms. All placebo-treated NJRM displayed acute dysentery symptoms within 24-36 hours of challenge. Two Travelan®-treated NJRM displayed dysentery symptoms and six animals remained healthy and symptom-free post challenge; resulting in 75% efficacy of prevention of shigellosis (p = 0.014). These results strongly indicate that Travelan® is functionally cross-reactive and an effective prophylactic for shigellosis. This has positive implications for the prophylactic use of Travelan® for protection against both ETEC and Shigella spp. diarrheal infections. Future refinement and expansion of pathogens recognized by HBC including Travelan® could revolutionize current management of gastrointestinal infections and outbreaks in travelers' including military, peacekeepers, humanitarian workers and in populations living in endemic regions of the world.
Collapse
Affiliation(s)
- Dilara Islam
- US Army Medical Directorate of the Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Nattaya Ruamsap
- US Army Medical Directorate of the Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Rawiwan Imerbsin
- US Army Medical Directorate of the Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Patchariya Khanijou
- US Army Medical Directorate of the Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Siriphan Gonwong
- US Army Medical Directorate of the Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Matthew D. Wegner
- US Army Medical Directorate of the Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Annette McVeigh
- Henry M. Jackson Foundation for Military Medicine (HJF), Bethesda, Maryland, United States of America
- Naval Medical Research Command (NMRC), Silver Spring, Maryland, United States of America
| | - Frédéric M. Poly
- Naval Medical Research Command (NMRC), Silver Spring, Maryland, United States of America
| | - John M. Crawford
- US Army Medical Directorate of the Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Brett E. Swierczewski
- US Army Medical Directorate of the Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Robert W. Kaminski
- Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland, United States of America
| | - Renee M. Laird
- Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland, United States of America
| |
Collapse
|
7
|
Khalil I, Anderson JD, Bagamian KH, Baqar S, Giersing B, Hausdorff WP, Marshall C, Porter CK, Walker RI, Bourgeois AL. Vaccine value profile for enterotoxigenic Escherichia coli (ETEC). Vaccine 2023; 41 Suppl 2:S95-S113. [PMID: 37951695 DOI: 10.1016/j.vaccine.2023.02.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/28/2022] [Accepted: 02/05/2023] [Indexed: 11/14/2023]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is one of the leading bacterial causes of diarrhoea, especially among children in low-resource settings, and travellers and military personnel from high-income countries. WHO's primary strategic goal for ETEC vaccine development is to develop a safe, effective, and affordable ETEC vaccine that reduces mortality and morbidity due to moderate-to-severe diarrhoeal disease in infants and children under 5 years of age in LMICs, as well as the long-term negative health impact on infant physical and cognitive development resulting from infection with this enteric pathogen. An effective ETEC vaccine will also likely reduce the need for antibiotic treatment and help limit the further emergence of antimicrobial resistance bacterial pathogens. The lead ETEC vaccine candidate, ETVAX, has shown field efficacy in travellers and has moved into field efficacy testing in LMIC infants and children. A Phase 3 efficacy study in LMIC infants is projected to start in 2024 and plans for a Phase 3 trial in travellers are under discussion with the U.S. FDA. Licensing for both travel and LMIC indications is projected to be feasible in the next 5-8 years. Given increasing recognition of its negative impact on child health and development in LMICs and predominance as the leading etiology of travellers' diarrhoea (TD), a standalone vaccine for ETEC is more cost-effective than vaccines targeting other TD pathogens, and a viable commercial market also exists. In contrast, combination of an ETEC vaccine with other vaccines for childhood pathogens in LMICs would maximize protection in a more cost-effective manner than a series of stand-alone vaccines. This 'Vaccine Value Profile' (VVP) for ETEC is intended to provide a high-level, holistic assessment of available data to inform the potential public health, economic and societal value of pipeline vaccines and vaccine-like products. This VVP was developed by a working group of subject matter experts from academia, non-profit organizations, public private partnerships, and multi-lateral organizations. All contributors have extensive expertise on various elements of the ETEC VVP and collectively aimed to identify current research and knowledge gaps. The VVP was developed using only existing and publicly available information.
Collapse
Affiliation(s)
- Ibrahim Khalil
- Department of Global Health, University of Washington, Seattle, WA 98195, USA.
| | - John D Anderson
- Bagamian Scientific Consulting, LLC, Gainesville, FL 32601, USA; Office of Health Affairs, West Virginia University, Morgantown, WV 26505, USA
| | - Karoun H Bagamian
- Bagamian Scientific Consulting, LLC, Gainesville, FL 32601, USA; Department of Environmental and Global Health, University of Florida, Gainesville, FL 32603, USA
| | - Shahida Baqar
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Birgitte Giersing
- Department of Immunization, Vaccines and Biologicals (IVB), World Health Organization (WHO), Geneva, Switzerland
| | - William P Hausdorff
- PATH, Center for Vaccine Innovation and Access, 455 Massachusetts Ave NW, Washington, DC 20001 USA; Faculty of Medicine, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Caroline Marshall
- Department of Immunization, Vaccines and Biologicals (IVB), World Health Organization (WHO), Geneva, Switzerland
| | - Chad K Porter
- Directorate for DoD Infectious Diseases Research, Naval Medical Research Command, Silver Spring, MD 20190, USA
| | - Richard I Walker
- PATH, Center for Vaccine Innovation and Access, 455 Massachusetts Ave NW, Washington, DC 20001 USA
| | - A Louis Bourgeois
- PATH, Center for Vaccine Innovation and Access, 455 Massachusetts Ave NW, Washington, DC 20001 USA
| |
Collapse
|
8
|
Rim S, Sakkestad ST, Zhou F, Gullaksen SE, Skavland J, Chauhan SK, Steinsland H, Hanevik K. Dynamics of circulating lymphocytes responding to human experimental enterotoxigenic Escherichia coli infection. Eur J Immunol 2023; 53:e2250254. [PMID: 37102399 DOI: 10.1002/eji.202250254] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/11/2023] [Accepted: 04/24/2023] [Indexed: 04/28/2023]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is an important cause of children's and travelers' diarrhea, with no licensed vaccine. This study aimed to explore the role of cellular immunity in protection against human ETEC infection. Nine volunteers were experimentally infected with ETEC, of which six developed diarrhea. Lymphocytes were collected from peripheral blood buffy coats, before and 3, 5, 6, 7, 10, and 28 days after dose ingestion, and 34 phenotypic and functional markers were examined by mass cytometry. Thirty-three cell populations, derived by manually merging 139 cell clusters from the X-shift unsupervised clustering algorithm, were analyzed. Initially, the diarrhea group responded with increased CD56dim CD16+ natural killer cells, dendritic cells tended to rise, and mucosal-associated invariant T cells decreased. On day 5-7, an increase in plasmablasts was paralleled by a consistent rise in CD4+ Th17-like effector memory and regulatory cell subsets. CD4+ Th17-like central memory cells peaked on day 10. All Th17-like cell populations showed increased expression of activation, gut-homing, and proliferation markers. Interestingly, in the nondiarrhea group, these same CD4+ Th17-like cell populations expanded earlier, normalizing around day 7. Earlier development of these CD4+ Th17-like cell populations in the nondiarrhea group may suggest a recall response and a potential role in controlling ETEC infections.
Collapse
Affiliation(s)
- Sehee Rim
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Sunniva T Sakkestad
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Fan Zhou
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Stein-Erik Gullaksen
- Department of Clinical Science, Centre of Cancer Biomarkers (CCBIO), University of Bergen, Bergen, Norway
- Hematology Section, Department of Internal Medicine, Helse Bergen, Bergen, Norway
| | - Jørn Skavland
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Sudhir K Chauhan
- Division of Cancer Medicine, Department of Cancer Immunology, Oslo University Hospital, Oslo, Norway
| | - Hans Steinsland
- Department of Global Public Health and Primary Care, Faculty of Medicine, Centre for Intervention Science in Maternal and Child Health (CISMAC), Centre for International Health, University of Bergen, Bergen, Norway
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Kurt Hanevik
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Medicine, Norwegian National Advisory Unit on Tropical Infectious Diseases, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
9
|
Khalid K, Poh CL. The Promising Potential of Reverse Vaccinology-Based Next-Generation Vaccine Development over Conventional Vaccines against Antibiotic-Resistant Bacteria. Vaccines (Basel) 2023; 11:1264. [PMID: 37515079 PMCID: PMC10385262 DOI: 10.3390/vaccines11071264] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
The clinical use of antibiotics has led to the emergence of multidrug-resistant (MDR) bacteria, leading to the current antibiotic resistance crisis. To address this issue, next-generation vaccines are being developed to prevent antimicrobial resistance caused by MDR bacteria. Traditional vaccine platforms, such as inactivated vaccines (IVs) and live attenuated vaccines (LAVs), were effective in preventing bacterial infections. However, they have shown reduced efficacy against emerging antibiotic-resistant bacteria, including MDR M. tuberculosis. Additionally, the large-scale production of LAVs and IVs requires the growth of live pathogenic microorganisms. A more promising approach for the accelerated development of vaccines against antibiotic-resistant bacteria involves the use of in silico immunoinformatics techniques and reverse vaccinology. The bioinformatics approach can identify highly conserved antigenic targets capable of providing broader protection against emerging drug-resistant bacteria. Multi-epitope vaccines, such as recombinant protein-, DNA-, or mRNA-based vaccines, which incorporate several antigenic targets, offer the potential for accelerated development timelines. This review evaluates the potential of next-generation vaccine development based on the reverse vaccinology approach and highlights the development of safe and immunogenic vaccines through relevant examples from successful preclinical and clinical studies.
Collapse
Affiliation(s)
- Kanwal Khalid
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Subang Jaya 47500, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Subang Jaya 47500, Malaysia
| |
Collapse
|
10
|
Strong Association between Diarrhea and Concentration of Enterotoxigenic Escherichia coli Strain TW10722 in Stools of Experimentally Infected Volunteers. Pathogens 2023; 12:pathogens12020283. [PMID: 36839555 PMCID: PMC9960819 DOI: 10.3390/pathogens12020283] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) strains are a major cause of diarrheal illness in children and travelers in low- and middle-income countries. When volunteers are infected with ETEC strains, as part of experimental infection studies, some do not develop diarrhea. To improve our understanding of how these volunteers are protected, we investigated the association between stool ETEC DNA concentration, as determined by quantitative PCR, and the development and severity of disease in 21 volunteers who had been experimentally infected with ETEC strain TW10722. We found a strong association between maximum stool ETEC DNA concentration and the development of diarrhea: all of the 11 volunteers who did not develop diarrhea had <0.99% TW10722-specific DNA in their stools throughout the follow-up period of up to 9 days, while all of the 10 volunteers who did develop diarrhea had maximum DNA concentrations of ≥0.99%. Most likely, these maximum stool TW10722 DNA concentrations reflect the level of intestinal colonization and the risk of experiencing diarrhea, thereby, seems to be directly dependent on the level of colonization. Thus, the development and availability of vaccines and other prophylactic measures, even if they only partially reduce colonization, could be important in the effort to reduce the burden of ETEC diarrhea.
Collapse
|
11
|
Porbahaie M, van den Belt M, Ulfman L, Ruijschop RMAJ, Lucas–van de Bos E, Hartog A, Lenz S, van Alen-Boerrigter IJ, Teodorowicz M, Savelkoul HFJ, Calame W, van Hoffen E, van Neerven RJJ, Kardinaal A. Low doses of diarrhoeagenic E. coli induce enhanced monocyte and mDC responses and prevent development of symptoms after homologous rechallenge. PLoS One 2023; 18:e0279626. [PMID: 36607972 PMCID: PMC9821474 DOI: 10.1371/journal.pone.0279626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 12/08/2022] [Indexed: 01/07/2023] Open
Abstract
The experimental challenge with attenuated enterotoxigenic E. coli strain E1392/75-2A prevents diarrhea upon a secondary challenge with the same bacteria. A dose-response pilot study was performed to investigate which immunological factors are associated with this protection. Healthy subjects were inoculated with increasing E. coli doses of 1E6-1E10 CFU, and three weeks later, all participants were rechallenged with the highest dose (1E10 CFU). Gastrointestinal discomfort symptoms were recorded, and stool and blood samples were analyzed. After the primary challenge, stool frequency, diarrhea symptom scores, and E. coli-specific serum IgG (IgG-CFA/II) titer increased in a dose-dependent manner. Fecal calprotectin and serum IgG-CFA/II response after primary challenge were delayed in the lower dose groups. Even though stool frequency after the secondary challenge was inversely related to the primary inoculation dose, all E. coli doses protected against clinical symptoms upon rechallenge. Ex vivo stimulation of PBMCs with E. coli just before the second challenge resulted in increased numbers of IL-6+/TNF-α+ monocytes and mDCs than before the primary challenge, without dose-dependency. These data demonstrate that primary E. coli infection with as few as 1E6 CFU protects against a high-dose secondary challenge with a homologous attenuated strain. Increased serum IgG-CFA/II levels and E. coli-induced mDC and monocyte responses after primary challenge suggest that protection against secondary E. coli challenges is associated with adaptive as well as innate immune responses.
Collapse
Affiliation(s)
- Mojtaba Porbahaie
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, The Netherlands
- * E-mail: (MP); (RJJN)
| | | | | | | | | | | | | | | | - Malgorzata Teodorowicz
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Huub F. J. Savelkoul
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, The Netherlands
| | | | | | - R. J. Joost van Neerven
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, The Netherlands
- FrieslandCampina, Amersfoort, The Netherlands
- * E-mail: (MP); (RJJN)
| | | |
Collapse
|
12
|
Choy RKM, Bourgeois AL, Ockenhouse CF, Walker RI, Sheets RL, Flores J. Controlled Human Infection Models To Accelerate Vaccine Development. Clin Microbiol Rev 2022; 35:e0000821. [PMID: 35862754 PMCID: PMC9491212 DOI: 10.1128/cmr.00008-21] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The timelines for developing vaccines against infectious diseases are lengthy, and often vaccines that reach the stage of large phase 3 field trials fail to provide the desired level of protective efficacy. The application of controlled human challenge models of infection and disease at the appropriate stages of development could accelerate development of candidate vaccines and, in fact, has done so successfully in some limited cases. Human challenge models could potentially be used to gather critical information on pathogenesis, inform strain selection for vaccines, explore cross-protective immunity, identify immune correlates of protection and mechanisms of protection induced by infection or evoked by candidate vaccines, guide decisions on appropriate trial endpoints, and evaluate vaccine efficacy. We prepared this report to motivate fellow scientists to exploit the potential capacity of controlled human challenge experiments to advance vaccine development. In this review, we considered available challenge models for 17 infectious diseases in the context of the public health importance of each disease, the diversity and pathogenesis of the causative organisms, the vaccine candidates under development, and each model's capacity to evaluate them and identify correlates of protective immunity. Our broad assessment indicated that human challenge models have not yet reached their full potential to support the development of vaccines against infectious diseases. On the basis of our review, however, we believe that describing an ideal challenge model is possible, as is further developing existing and future challenge models.
Collapse
Affiliation(s)
- Robert K. M. Choy
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | - A. Louis Bourgeois
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Richard I. Walker
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Jorge Flores
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| |
Collapse
|
13
|
Rocha-Melogno L, Crank KC, Ginn O, Bergin MH, Brown J, Gray GC, Hamilton KA, Bibby K, Deshusses MA. Quantitative microbial risk assessment of outdoor aerosolized pathogens in cities with poor sanitation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 827:154233. [PMID: 35245543 DOI: 10.1016/j.scitotenv.2022.154233] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/08/2022] [Accepted: 02/25/2022] [Indexed: 06/14/2023]
Abstract
The aeromicrobiological transmission pathway of enteric pathogens in places with unsafe sanitation services is poorly understood. In an attempt to partly fill this knowledge gap, we assessed the potential public health impact of bioaerosols near open waste canals (OWCs) using Quantitative Microbial Risk Assessment (QMRA). We used data acquired in La Paz, Bolivia to characterize the risk of disease that aerosolized enteric pathogens may pose through food, fomites and inhalation (all followed by ingestion). Three reference pathogens were selected to conduct the assessment: enterotoxigenic Escherichia coli (ETEC), Shigella flexneri, and Campylobacter jejuni. Inhalation followed by ingestion had the highest median infection risk per event i.e. 3 × 10-5 (3 infections for every 100,000 exposures), compared to contaminated food e.g. 5 × 10-6 and fomites e.g. 2 × 10-7, all for C. jejuni infections. Our sensitivity analysis showed that bacterial fluxes from the air were the most influential factor on risk. Our results suggest that fecal bacterial aerosols from OWCs present non-negligible risks of infection in La Paz, with median annual infection risks by C. jejuni being 18 (food), and 100 (inhalation) times greater than the EPA's standard for drinking water (1 × 10-4). We included two of the QMRA models presented here in a novel web application we developed for user-specified application in different contexts.
Collapse
Affiliation(s)
- Lucas Rocha-Melogno
- Department of Civil and Environmental Engineering, Duke University, Durham, NC 27708, United States; Duke Global Health Institute, Duke University, Durham, NC 27710, United States; ICF, 2635 Meridian Parkway Suite 200, Durham, NC 27713, United States
| | - Katherine C Crank
- Department of Civil and Environmental Engineering and Earth Sciences, University of Notre Dame, IN 46556, United States
| | - Olivia Ginn
- School of Civil and Environmental Engineering, Georgia Institute of Technology, Atlanta, GA 30332, United States
| | - Michael H Bergin
- Department of Civil and Environmental Engineering, Duke University, Durham, NC 27708, United States
| | - Joe Brown
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, United States
| | - Gregory C Gray
- Duke Global Health Institute, Duke University, Durham, NC 27710, United States; Division of Infectious Diseases, Duke University School of Medicine, Durham, NC 27710, United States; Global Health Research Center, Duke-Kunshan University, Kunshan, China; Emerging Infectious Diseases Program, Duke-NUS Medical School, Singapore, Singapore; Division of Infectious Diseases, University of Texas Medical Branch (UTMB), Galveston, TX 77555, United States
| | - Kerry A Hamilton
- School of Sustainable Engineering and the Built Environment, Arizona State University, 770 S College Ave, Tempe, AZ 85281, United States; The Biodesign Institute Center for Environmental Health Engineering, Arizona State University, 1001 S McAllister Ave, Tempe, AZ 85281, United States
| | - Kyle Bibby
- Department of Civil and Environmental Engineering and Earth Sciences, University of Notre Dame, IN 46556, United States
| | - Marc A Deshusses
- Department of Civil and Environmental Engineering, Duke University, Durham, NC 27708, United States; Duke Global Health Institute, Duke University, Durham, NC 27710, United States.
| |
Collapse
|
14
|
Sheikh A, Wangdi T, Vickers TJ, Aaron B, Palmer M, Miller MJ, Kim S, Herring C, Simoes R, Crainic JA, Gildersleeve JC, van der Post S, Hansson GC, Fleckenstein JM. Enterotoxigenic Escherichia coli Degrades the Host MUC2 Mucin Barrier To Facilitate Critical Pathogen-Enterocyte Interactions in Human Small Intestine. Infect Immun 2022; 90:e0057221. [PMID: 34807735 PMCID: PMC8853678 DOI: 10.1128/iai.00572-21] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 11/12/2021] [Indexed: 02/08/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) isolates are genetically diverse pathological variants of E. coli defined by the production of heat-labile (LT) and/or heat-stable (ST) toxins. ETEC strains are estimated to cause hundreds of millions of cases of diarrheal illness annually. However, it is not clear that all strains are equally equipped to cause disease, and asymptomatic colonization with ETEC is common in low- to middle-income regions lacking basic sanitation and clean water where ETEC are ubiquitous. Recent molecular epidemiology studies have revealed a significant association between strains that produce EatA, a secreted autotransporter protein, and the development of symptomatic infection. Here, we demonstrate that LT stimulates production of MUC2 mucin by goblet cells in human small intestine, enhancing the protective barrier between pathogens and enterocytes. In contrast, using explants of human small intestine as well as small intestinal enteroids, we show that EatA counters this host defense by engaging and degrading the MUC2 mucin barrier to promote bacterial access to target enterocytes and ultimately toxin delivery, suggesting that EatA plays a crucial role in the molecular pathogenesis of ETEC. These findings may inform novel approaches to prevention of acute diarrheal illness as well as the sequelae associated with ETEC and other pathogens that rely on EatA and similar proteases for efficient interaction with their human hosts.
Collapse
Affiliation(s)
- Alaullah Sheikh
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Tamding Wangdi
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Tim J. Vickers
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Bailey Aaron
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Margot Palmer
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Mark J. Miller
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Seonyoung Kim
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Cassandra Herring
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Rita Simoes
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Jennifer A. Crainic
- Center for Cancer Research, Chemical Biology Laboratory, National Cancer Institute, Fredrick, Maryland, USA
| | - Jeffrey C. Gildersleeve
- Center for Cancer Research, Chemical Biology Laboratory, National Cancer Institute, Fredrick, Maryland, USA
| | - Sjoerd van der Post
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Gunnar C. Hansson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - James M. Fleckenstein
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
- Medicine Service, Veterans Affairs Medical Center, Saint Louis, Missouri, USA
| |
Collapse
|
15
|
Connor S, Velagic M, Zhang X, Johura FT, Chowdhury G, Mukhopadhyay AK, Dutta S, Alam M, Sack DA, Wierzba TF, Chakraborty S. Evaluation of a simple, rapid and field-adapted diagnostic assay for enterotoxigenic E. coli and Shigella. PLoS Negl Trop Dis 2022; 16:e0010192. [PMID: 35130310 PMCID: PMC8853640 DOI: 10.1371/journal.pntd.0010192] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 02/17/2022] [Accepted: 01/21/2022] [Indexed: 11/19/2022] Open
Abstract
Understanding the global burden of enterotoxigenic E. coli (ETEC) and Shigella diarrhea as well as estimating the cost effectiveness of vaccines to control these two significant pathogens have been hindered by the lack of a diagnostic test that is rapid, simple, sensitive, and can be applied to the endemic countries. We previously developed a simple and rapid assay, Rapid Loop mediated isothermal amplification based Diagnostic Test (RLDT) for the detection of ETEC and Shigella spp. (Shigella). In this study, the RLDT assay was evaluated in comparison with quantitative PCR (qPCR), culture and conventional PCR for the detection of ETEC and Shigella. This validation was performed using previously collected stool samples from endemic countries, from the travelers to the endemic countries, as well as samples from a controlled human infection model study of ETEC. The performance of RLDT from dried stool spots was also validated. RLDT resulted in excellent sensitivity and specificity compared to qPCR (99% and 99.2% respectively) ranging from 92.3 to 100% for the individual toxin genes of ETEC and 100% for Shigella. Culture was less sensitive compared to RLDT. No significant differences were noted in the performance of RLDT using samples from various sources or stool samples from moderate to severe diarrhea or asymptomatic infections. RLDT performed equally well in detection of ETEC and Shigella from the dried stool samples on filter papers. This study established that RLDT is sufficiently sensitive and specific to be used as a simple and rapid diagnostic assay to detect ETEC and Shigella in endemic countries to determine disease burden of these pathogens in the national and subnational levels. This information will be important to guide public health and policy makers to prioritize resources for accelerating the development and introduction of effective preventative and/or treatment interventions against these enteric infections. Enterotoxigenic E. coli (ETEC) and Shigella spp (Shigella) causes significant global morbidity and mortality, especially in low-and middle-income countries (LMICs). Since culture methods to detect Shigella are not sensitive, and the methods used to detect ETEC have not been feasible outside of specialized, well-equipped laboratories, the true burden of these pathogens at national and sub-national levels are mostly not available. Morbidity and mortality estimates, for these two pathogens are crucial to assess their relative public health importance in LMICs. We developed a simple and rapid diagnostic assay called the RLDT (Rapid Loop-mediated isothermal amplification based Diagnostic Test) for detection of ETEC and Shigella. In this study we evaluated RLDT compared to other currently available assays using previously collected stool samples. Our data showed that the RLDT assay exhibited high sensitivity and specificity for detection of ETEC and Shigella, with its result available within 50 minutes. The sensitivity of RLDT was higher than culture for these pathogens. We conclude that RLDT could be used as a rapid and simple diagnostic test to determine the burden of ETEC and Shigella in LMICs as well as in clinical vaccine trials of these pathogens.
Collapse
Affiliation(s)
- Sean Connor
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Mirza Velagic
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Xueyan Zhang
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Fatema-Tuz Johura
- icddr,b, Formerly International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Goutam Chowdhury
- ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | | | - Shanta Dutta
- ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Munirul Alam
- icddr,b, Formerly International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - David A. Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Thomas F. Wierzba
- Wake Forest School of Medicine, Winston Salem, North Carolina, United States of America
| | - Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
16
|
Chakraborty S, Connor S, Velagic M. Development of a simple, rapid, and sensitive diagnostic assay for enterotoxigenic E. coli and Shigella spp applicable to endemic countries. PLoS Negl Trop Dis 2022; 16:e0010180. [PMID: 35089927 PMCID: PMC8827434 DOI: 10.1371/journal.pntd.0010180] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 02/09/2022] [Accepted: 01/18/2022] [Indexed: 11/18/2022] Open
Abstract
Enterotoxigenic E. coli (ETEC) and Shigella spp (Shigella) are complex pathogens. The diagnostic assays currently used to detect these pathogens are elaborate or complicated, which make them difficult to apply in resource poor settings where these diseases are endemic. The culture methods used to detect Shigella are not sensitive, and the methods used to detect ETEC are only available in a few research labs. To address this gap, we developed a rapid and simple diagnostic assay–"Rapid LAMP based Diagnostic Test (RLDT)." The six minutes sample preparation method directly from the fecal samples with lyophilized reaction strips and using established Loop-mediated Isothermal Amplification (LAMP) platform, ETEC [heat labile toxin (LT) and heat stable toxins (STh, and STp) genes] and Shigella (ipaH gene) detection was made simple, rapid (<50 minutes), and inexpensive. This assay is cold chain and electricity free. Moreover, RLDT requires minimal equipment. To avoid any end user’s bias, a battery-operated, handheld reader was used to read the RLDT results. The results can be read as positive/negative or as real time amplification depending on the end user’s need. The performance specifications of the RLDT assay, including analytical sensitivity and specificity, were evaluated using fecal samples spiked with ETEC and Shigella strains. The limit of detection was ~105 CFU/gm of stool for LT, STh, and STp and ~104 CFU/gm of stool for the ipaH gene, which corresponds to about 23 CFU and 1 CFU respectively for ETEC and Shigella per 25uL reaction within 40 minutes. The RLDT assay from stool collection to result is simple, and rapid and at the same time sufficiently sensitive. RLDT has the potential to be applied in resource poor endemic settings for the rapid diagnosis of ETEC and Shigella. Enterotoxigenic E. coli and Shigella are the leading causes of moderate to severe diarrhea in the low-and middle-income countries (LMICs). A critical constraint to determine the ETEC and Shigella disease burden at the country or sub-national level, is the complex diagnostic methods currently required for detecting these pathogens. These methods are neither sufficiently sensitive nor standardized and are not feasible in the resource poor settings where these infections occur most commonly. We developed a simple and rapid diagnostic assay called "Rapid Loop-mediated isothermal amplification based Diagnostic Test (RLDT)" for the detection of these pathogens in low-resource settings. Using RLDT, ETEC and Shigella were detected directly from the stool, in less than 1 hour with minimal hands-on time. The assay does not require maintaining a cold chain and is electricity-free. Being rapid, simple, and sensitive, RLDT can be scaled up and is appropriate to apply in the LMICs where ETEC and Shigella diarrhea are endemic.
Collapse
Affiliation(s)
- Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- * E-mail:
| | - Sean Connor
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Mirza Velagic
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
17
|
McMurry TL, McQuade ETR, Liu J, Kang G, Kosek MN, Lima AAM, Bessong PO, Samie A, Haque R, Mduma ER, Leite JP, Bodhidatta L, Iqbal NT, Page N, Kiwelu I, Bhutta ZA, Ahmed T, Houpt ER, Platts-Mills JA. Duration of Postdiarrheal Enteric Pathogen Carriage in Young Children in Low-resource Settings. Clin Infect Dis 2021; 72:e806-e814. [PMID: 33033835 PMCID: PMC8315229 DOI: 10.1093/cid/ciaa1528] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Indexed: 02/07/2023] Open
Abstract
Background Prolonged enteropathogen shedding after diarrhea complicates the identification of etiology in subsequent episodes and is an important driver of pathogen transmission. A standardized approach has not been applied to estimate the duration of shedding for a wide range of pathogens. Methods We used a multisite birth cohort of children 0–24 months of age from whom diarrheal and monthly nondiarrheal stools were previously tested by quantitative polymerase chain reaction for 29 enteropathogens. We modeled the probability of detection of the etiologic pathogen before and after diarrhea using a log-normal accelerated failure time survival model and estimated the median duration of pathogen carriage as well as differences in subclinical pathogen carriage 60 days after diarrhea onset in comparison to a prediarrhea baseline. Results We analyzed 3247 etiologic episodes of diarrhea for the 9 pathogens with the highest attributable burdens of diarrhea. The median duration of postdiarrheal carriage varied widely by pathogen, from about 1 week for rotavirus (median, 8.1 days [95% confidence interval {CI}, 6.2–9.6]) to >1 month for Cryptosporidium (39.5 days [95% CI, 30.6–49.0]). The largest increases in subclinical pathogen carriage before and after diarrhea were seen for Cryptosporidium (prevalence difference between 30 days prior and 60 days after diarrhea onset, 0.30 [95% CI, .23–.39]) and Shigella (prevalence difference, 0.21 [95% CI, .16–.27]). Conclusions Postdiarrheal shedding was widely variable between pathogens, with strikingly prolonged shedding seen for Cryptosporidium and Shigella. Targeted antimicrobial therapy and vaccination for these pathogens may have a relatively large impact on transmission.
Collapse
Affiliation(s)
- Timothy L McMurry
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Elizabeth T Rogawski McQuade
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA.,Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Jie Liu
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | | | - Margaret N Kosek
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA.,Asociación Benéfica PRISMA, Iquitos, Peru
| | | | | | | | - Rashidul Haque
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | | | | | | | | | - Nicola Page
- National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Ireen Kiwelu
- Kilimanjaro Clinical Research Institute, Moshi, Tanzania
| | | | - Tahmeed Ahmed
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Eric R Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - James A Platts-Mills
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
18
|
Fabri LV, Azzopardi KI, Osowicki J, Frost HR, Smeesters PR, Steer AC. An emm-type specific qPCR to track bacterial load during experimental human Streptococcus pyogenes pharyngitis. BMC Infect Dis 2021; 21:463. [PMID: 34020607 PMCID: PMC8138111 DOI: 10.1186/s12879-021-06173-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 05/12/2021] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Streptococcus pyogenes causes a profound global burden of morbidity and mortality across its diverse clinical spectrum. To support a new controlled human infection ('challenge') model seeking to accelerate S. pyogenes vaccine development, we aimed to develop an accurate and reliable molecular method for quantifying bacterial load from pharyngeal swabs collected during experimental human pharyngitis. METHODS Combined sequential RNA + DNA extraction from throat swabs was compared to traditional separate RNA-only and DNA-only extractions. An emm-type specific qPCR was developed to detect the emm75 challenge strain. Results from the qPCR were compared to culture, using throat swab samples collected in a human challenge study. RESULTS The qPCR was 100% specific for the emm75 challenge strain when tested against a panel of S. pyogenes emm-types and other respiratory pathogens. Combined RNA + DNA extraction had similar yield to traditional separate extractions. The combined extraction method and emm75 qPCR had 98.8% sensitivity compared to culture for throat swabs collected from challenge study participants. CONCLUSIONS We have developed a reliable molecular method for measuring S. pyogenes bacterial load from throat swabs collected in a controlled human infection model of S. pyogenes pharyngitis. TRIAL REGISTRATION NCT03361163 on 4th December 2017.
Collapse
Affiliation(s)
- Loraine V Fabri
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, Université Libre de Bruxelles, Brussels, Belgium
| | - Kristy I Azzopardi
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Joshua Osowicki
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.
- Infectious Diseases Unit, Department of General Medicine, The Royal Children's Hospital Melbourne, Melbourne, Victoria, Australia.
| | - Hannah R Frost
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Pierre R Smeesters
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, Université Libre de Bruxelles, Brussels, Belgium
- Academic Children Hospital Queen Fabiola, Université Libre de Bruxelles, Brussels, Belgium
- Molecular Bacteriology Laboratory, Université Libre de Bruxelles, Brussels, Belgium
| | - Andrew C Steer
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Infectious Diseases Unit, Department of General Medicine, The Royal Children's Hospital Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
19
|
van Hoffen E, Mercenier A, Vidal K, Benyacoub J, Schloesser J, Kardinaal A, Lucas-van de Bos E, van Alen I, Roggero I, Duintjer K, Berendts A, Albers R, Kleerebezem M, Ten Bruggencate S. Characterization of the pathophysiological determinants of diarrheagenic Escherichia coli infection using a challenge model in healthy adults. Sci Rep 2021; 11:6060. [PMID: 33723346 PMCID: PMC7960709 DOI: 10.1038/s41598-021-85161-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/15/2021] [Indexed: 11/09/2022] Open
Abstract
An experimental human challenge model with an attenuated diarrheagenic Escherichia coli (E. coli) strain has been used in food intervention studies aimed to increase resistance to E. coli infection. This study was designed to refine and expand this challenge model. In a double-blind study, healthy male subjects were orally challenged with 1E10 or 5E10 colony-forming units (CFU) of E. coli strain E1392/75-2A. Three weeks later, subjects were rechallenged with 1E10 CFU of E. coli. Before and after both challenges, clinical symptoms and infection- and immune-related biomarkers were analyzed. Subset analysis was performed on clinically high- and low-responders. Regardless of inoculation dose, the first challenge induced clinical symptoms for 2-3 days. In blood, neutrophils, CRP, CXCL10, and CFA/II-specific IgG were induced, and in feces calprotectin and CFA/II-specific IgA. Despite clinical differences between high- and low-responders, infection and immune biomarkers did not differ. The first inoculation induced protection at the second challenge, with a minor clinical response, and no change in biomarkers. The refined study design resulted in a larger dynamic range of symptoms, and identification of biomarkers induced by a challenge with the attenuated E. coli strain E1392/75-2A, which is of value for future intervention studies. Addition of a second inoculation allows to study the protective response induced by a primary infection.Clinicaltrials.gov registration: NCT02541695 (04/09/2015).
Collapse
Affiliation(s)
- Els van Hoffen
- Department of Nutrition and Health, NIZO, PO Box 20, 6710 BA, Ede, the Netherlands
| | - Annick Mercenier
- Nestlé Institute of Health Sciences, Gastrointestinal Health, Nestlé Research, Lausanne, Switzerland.,NutriLeads, Wageningen, The Netherlands
| | - Karine Vidal
- Nestlé Institute of Health Sciences, Gastrointestinal Health, Nestlé Research, Lausanne, Switzerland
| | - Jalil Benyacoub
- Nestlé Institute of Health Sciences, Gastrointestinal Health, Nestlé Research, Lausanne, Switzerland
| | - Joyce Schloesser
- Department of Nutrition and Health, NIZO, PO Box 20, 6710 BA, Ede, the Netherlands.
| | - Alwine Kardinaal
- Department of Nutrition and Health, NIZO, PO Box 20, 6710 BA, Ede, the Netherlands
| | | | - Ingrid van Alen
- Department of Nutrition and Health, NIZO, PO Box 20, 6710 BA, Ede, the Netherlands
| | - Iris Roggero
- Nestlé Institute of Health Sciences, Gastrointestinal Health, Nestlé Research, Lausanne, Switzerland
| | - Kim Duintjer
- Department of Nutrition and Health, NIZO, PO Box 20, 6710 BA, Ede, the Netherlands.,Host Microbe Interactomics Group, Department of Animal Sciences, Wageningen University and Research, Wageningen, the Netherlands
| | - Anneke Berendts
- Department of Nutrition and Health, NIZO, PO Box 20, 6710 BA, Ede, the Netherlands
| | | | - Michiel Kleerebezem
- Host Microbe Interactomics Group, Department of Animal Sciences, Wageningen University and Research, Wageningen, the Netherlands
| | | |
Collapse
|
20
|
Establishment and Validation of Pathogenic CS17 + and CS19 + Enterotoxigenic Escherichia coli Challenge Models in the New World Primate Aotus nancymaae. Infect Immun 2021; 89:IAI.00479-20. [PMID: 33288648 DOI: 10.1128/iai.00479-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/23/2020] [Indexed: 11/20/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a common cause of diarrheal illness in the military, travelers, and children living in low- to middle-income countries. Increased antibiotic resistance, the absence of a licensed vaccine, and the lack of broadly practical therapeutics perpetuate the significant health and financial burden resulting from ETEC infection. A critical step in the evaluation of vaccines and therapeutics is preclinical screening in a relevant animal disease model that closely replicates human disease. We previously developed a diarrheal model of class 5a colonization factor (CF) CFA/I-expressing ETEC in the New World owl monkey species Aotus nancymaae using ETEC strain H10407. In order to broaden the use of the model, we report here on the development of A. nancymaae models of ETEC expressing the class 5b CFs CS17 and CS19 with strains LSN03-016011/A and WS0115A, respectively. For both models, we observed diarrheal attack rates of ≥80% after oral inoculation with 5 × 1011 CFU of bacteria. These models will aid in assessing the efficacy of future ETEC vaccine candidates and therapeutics.
Collapse
|
21
|
Brubaker J, Zhang X, Bourgeois AL, Harro C, Sack DA, Chakraborty S. Intestinal and systemic inflammation induced by symptomatic and asymptomatic enterotoxigenic E. coli infection and impact on intestinal colonization and ETEC specific immune responses in an experimental human challenge model. Gut Microbes 2021; 13:1-13. [PMID: 33645430 PMCID: PMC7919917 DOI: 10.1080/19490976.2021.1891852] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 02/02/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
Recent studies have gained a better appreciation of the potential impacts of enteric infections beyond symptomatic diarrhea. It is recognized that infections by several enteropathogens could be associated with growth deficits in children and intestinal and systemic inflammation may play an important underlying role. With enterotoxigenic E. coli (ETEC) being one of the leading causes of diarrhea among children in the developing world and important contributor to stunting, a better understanding of the impact of ETEC infection beyond diarrhea is timely and greatly needed. To address this, we evaluated if ETEC infection induces intestinal and systemic inflammation and its impact on colonization and immune responses to ETEC vaccine-specific antigens in a dose descending experimental human challenge model using ETEC strain H10407. This study demonstrates that the concentrations of myeloperoxidase (MPO) in stool and intestinal fatty acid-binding protein (an indicator of compromised intestinal epithelial integrity) in serum, significantly increased following ETEC infection in both diarrhea and asymptomatic cases and the magnitudes and kinetics of MPO are dose and clinical outcome dependent. Cytokines IL-17A and IFN-γ were significantly increased in serum post-ETEC challenge. In addition, higher pre-challenge concentrations of cytokines IL-10 and GM-CSF were associated with protection from ETEC diarrhea. Interestingly, higher MPO concentrations were associated with higher intestinal colonization of ETEC and lower seroconversions of colonization factor I antigen, but the reverse was noted for seroconversions to heat-labile toxin B-subunit. Together this study has important implications for understanding the acute and long-term negative health outcomes associated with ETEC infection.
Collapse
Affiliation(s)
- Jessica Brubaker
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Xueyan Zhang
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - A. Louis Bourgeois
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
- PATH, Center for Vaccine Innovation and Access, Washington, DC, 20001, USA
| | - Clayton Harro
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - David A Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
22
|
Talaat KR, Porter CK, Jaep KM, Duplessis CA, Gutierrez RL, Maciel M, Adjoodani B, Feijoo B, Chakraborty S, Brubaker J, Trop SA, Riddle MS, Joseph SS, Bourgeois AL, Prouty MG. Refinement of the CS6-expressing enterotoxigenic Escherichia coli strain B7A human challenge model: A randomized trial. PLoS One 2020; 15:e0239888. [PMID: 33264302 PMCID: PMC7710093 DOI: 10.1371/journal.pone.0239888] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 09/01/2020] [Indexed: 12/29/2022] Open
Abstract
Background Human challenge models for enterotoxigenic Escherichia coli (ETEC) facilitate vaccine down-selection. The B7A (O148:H28 CS6+LT+ST+) strain is important for vaccine development. We sought to refine the B7A model by identifying a dose and fasting regimen consistently inducing moderate-severe diarrhea. Methods An initial cohort of 28 subjects was randomized (1:1:1:1) to receive B7A following an overnight fast at doses of 108 or 109 colony forming units (cfu) or a 90-minute fast at doses of 109 or 1010 cfu. A second cohort included naïve and rechallenged subjects who had moderate-severe diarrhea and were given the target regimen. Immune responses to important ETEC antigens were assessed. Results Among subjects receiving 108 cfu of B7A, overnight fast, or 109 cfu, 90-minute fast, 42.9% (3/7) had moderate-severe diarrhea. Higher attack rates (71.4%; 5/7) occurred in subjects receiving 109 cfu, overnight fast, or 1010 cfu, 90-minute fast. Upon rechallenge with 109 cfu of B7A, overnight fast, 5/11 (45.5%) had moderate-severe diarrhea; the attack rate among concurrently challenge naïve subjects was 57.9% (11/19). Anti-CS6, O148 LPS and LT responses were modest across all groups. Conclusions An overnight fast enabled a reduction in the B7A inoculum dose; however, the attack rate was inconsistent and protection upon rechallenge was minimal.
Collapse
Affiliation(s)
- Kawsar R. Talaat
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States of America
- * E-mail:
| | - Chad K. Porter
- Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Kayla M. Jaep
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | | | | | - Milton Maciel
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Brittany Adjoodani
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Brittany Feijoo
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Subhra Chakraborty
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Jessica Brubaker
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Stefanie A. Trop
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Mark S. Riddle
- Naval Medical Research Center, Silver Spring, MD, United States of America
- Uniformed Services University of the Health Sciences, Bethesda, MD, United States of America
| | | | - A. Louis Bourgeois
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States of America
- PATH, Washington, DC, United States of America
| | - Michael G. Prouty
- Naval Medical Research Center, Silver Spring, MD, United States of America
| |
Collapse
|
23
|
Talaat KR, Porter CK, Bourgeois AL, Lee TK, Duplessis CA, Maciel M, Gutierrez RL, DeNearing B, Adjoodani B, Adkinson R, Testa KJ, Feijoo B, Alcala AN, Brubaker J, Beselman A, Chakraborty S, Sack D, Halpern J, Trop S, Wu H, Jiao J, Sullivan E, Riddle MS, Joseph SS, Poole ST, Prouty MG. Oral delivery of Hyperimmune bovine serum antibodies against CS6-expressing enterotoxigenic Escherichia coli as a prophylactic against diarrhea. Gut Microbes 2020; 12:1732852. [PMID: 32167011 PMCID: PMC7524165 DOI: 10.1080/19490976.2020.1732852] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND . Oral administration of bovine antibodies active against enterotoxigenic Escherichia coli (ETEC) have demonstrated safety and efficacy against diarrhea in human challenge trials. The efficacy of bovine serum immunoglobulins (BSIgG) against recombinant colonization factor CS6 or whole cell ETEC strain B7A was assessed against challenge with the CS6-expressing B7A. METHODS . This was a randomized, double-blind, placebo-controlled trial in which healthy adults received oral hyperimmune BSIgG anti-CS6, anti-B7A whole cell killed or non-hyperimmune BSIgG (placebo) in a 1:1:1 ratio then challenged with ETEC B7A. Two days pre-challenge, volunteers began a thrice daily, seven day course of immunoprophylaxis. On day 3, subjects received 1 × 1010 CFUs of B7A. Subjects were observed for safety and the primary endpoint of moderate-severe diarrhea (MSD). RESULTS . A total of 59 volunteers received product and underwent ETEC challenge. The BSIgG products were well-tolerated across all subjects. Upon challenge, 14/20 (70%) placebo recipients developed MSD, compared to 12/19 (63%; p = .74) receiving anti-CS6 BSIgG and 7/20 (35%; p = .06) receiving anti-B7A BSIgG. Immune responses to the ETEC infection were modest across all groups. CONCLUSIONS . Bovine-derived serum antibodies appear safe and well tolerated. Antibodies derived from cattle immunized with whole cell B7A provided 50% protection against MSD following B7A challenge; however, no protection was observed in subjects receiving serum antibodies targeting CS6. The lack of observed efficacy in this group may be due to low CS6 surface expression on B7A, the high dose challenge inoculum and/or the use of serum derived antibodies versus colostrum-derived antibodies.
Collapse
Affiliation(s)
- KR Talaat
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - CK Porter
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, USA,CONTACT CK Porter Naval Medical Research Center, Silver Spring, MD, USA
| | - AL Bourgeois
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - TK Lee
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, USA
| | - CA Duplessis
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, USA
| | - M Maciel
- The Henry M. Jackson Foundation, Bethesda, MD, USA
| | - RL Gutierrez
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, USA
| | - B DeNearing
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - B Adjoodani
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - R Adkinson
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - KJ Testa
- The Henry M. Jackson Foundation, Bethesda, MD, USA
| | - B Feijoo
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - AN Alcala
- The Henry M. Jackson Foundation, Bethesda, MD, USA
| | - J Brubaker
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - A Beselman
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - S Chakraborty
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - D Sack
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - J Halpern
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - S Trop
- The Henry M. Jackson Foundation, Bethesda, MD, USA
| | - H Wu
- SAB Biotherapeutics Inc, Sioux Falls, SD, USA
| | - J Jiao
- SAB Biotherapeutics Inc, Sioux Falls, SD, USA
| | - E Sullivan
- SAB Biotherapeutics Inc, Sioux Falls, SD, USA
| | - MS Riddle
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, USA
| | - SS Joseph
- The Henry M. Jackson Foundation, Bethesda, MD, USA
| | - ST Poole
- The Henry M. Jackson Foundation, Bethesda, MD, USA
| | - MG Prouty
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, USA
| |
Collapse
|
24
|
Gibani MM, Jin C, Shrestha S, Moore M, Norman L, Voysey M, Jones E, Blackwell L, Thomaides-Brears H, Hill J, Blohmke CJ, Dobinson HC, Baker P, Jones C, Campbell D, Mujadidi YF, Plested E, Preciado-Llanes L, Napolitani G, Simmons A, Gordon MA, Angus B, Darton TC, Cerundulo V, Pollard AJ. Homologous and heterologous re-challenge with Salmonella Typhi and Salmonella Paratyphi A in a randomised controlled human infection model. PLoS Negl Trop Dis 2020; 14:e0008783. [PMID: 33079959 PMCID: PMC7598925 DOI: 10.1371/journal.pntd.0008783] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/30/2020] [Accepted: 09/08/2020] [Indexed: 11/19/2022] Open
Abstract
Enteric fever is a systemic infection caused by Salmonella Typhi or Paratyphi A. In many endemic areas, these serovars co-circulate and can cause multiple infection-episodes in childhood. Prior exposure is thought to confer partial, but incomplete, protection against subsequent attacks of enteric fever. Empirical data to support this hypothesis are limited, and there are few studies describing the occurrence of heterologous-protection between these closely related serovars. We performed a challenge-re-challenge study using a controlled human infection model (CHIM) to investigate the extent of infection-derived immunity to Salmonella Typhi or Paratyphi A infection. We recruited healthy volunteers into two groups: naïve volunteers with no prior exposure to Salmonella Typhi/Paratyphi A and volunteers previously-exposed to Salmonella Typhi or Paratyphi A in earlier CHIM studies. Within each group, participants were randomised 1:1 to oral challenge with either Salmonella Typhi (104 CFU) or Paratyphi A (103 CFU). The primary objective was to compare the attack rate between naïve and previously challenged individuals, defined as the proportion of participants per group meeting the diagnostic criteria of temperature of ≥38°C persisting for ≥12 hours and/or S. Typhi/Paratyphi bacteraemia up to day 14 post challenge. The attack-rate in participants who underwent homologous re-challenge with Salmonella Typhi was reduced compared with challenged naïve controls, although this reduction was not statistically significant (12/27[44%] vs. 12/19[63%]; Relative risk 0.70; 95% CI 0.41-1.21; p = 0.24). Homologous re-challenge with Salmonella Paratyphi A also resulted in a lower attack-rate than was seen in challenged naïve controls (3/12[25%] vs. 10/18[56%]; RR0.45; 95% CI 0.16-1.30; p = 0.14). Evidence of protection was supported by a post hoc analysis in which previous exposure was associated with an approximately 36% and 57% reduced risk of typhoid or paratyphoid disease respectively on re-challenge. Individuals who did not develop enteric fever on primary exposure were significantly more likely to be protected on re-challenge, compared with individuals who developed disease on primary exposure. Heterologous re-challenge with Salmonella Typhi or Salmonella Paratyphi A was not associated with a reduced attack rate following challenge. Within the context of the model, prior exposure was not associated with reduced disease severity, altered microbiological profile or boosting of humoral immune responses. We conclude that prior Salmonella Typhi and Paratyphi A exposure may confer partial but incomplete protection against subsequent infection, but with a comparable clinical and microbiological phenotype. There is no demonstrable cross-protection between these serovars, consistent with the co-circulation of Salmonella Typhi and Paratyphi A. Collectively, these data are consistent with surveillance and modelling studies that indicate multiple infections can occur in high transmission settings, supporting the need for vaccines to reduce the burden of disease in childhood and achieve disease control. Trial registration NCT02192008; clinicaltrials.gov.
Collapse
Affiliation(s)
- Malick M. Gibani
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
- Department of Infectious Diseases, Imperial College London, United Kingdom
| | - Celina Jin
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Sonu Shrestha
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Maria Moore
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Lily Norman
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Merryn Voysey
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Elizabeth Jones
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Luke Blackwell
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Helena Thomaides-Brears
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Jennifer Hill
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Christoph J. Blohmke
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Hazel C. Dobinson
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Philip Baker
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Claire Jones
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Danielle Campbell
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Yama F. Mujadidi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Emma Plested
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Lorena Preciado-Llanes
- Institute for Infection and Global Health, University of Liverpool, United Kingdom
- Medical Research Council Human Immunology Unit, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| | - Giorgio Napolitani
- Medical Research Council Human Immunology Unit, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| | - Alison Simmons
- Medical Research Council Human Immunology Unit, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| | - Melita A. Gordon
- Institute for Infection and Global Health, University of Liverpool, United Kingdom
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Brian Angus
- Nuffield Department of Medicine, University of Oxford, United Kingdom
| | - Thomas C. Darton
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Vincenzo Cerundulo
- Medical Research Council Human Immunology Unit, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| | - Andrew J. Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| |
Collapse
|
25
|
Riaz S, Steinsland H, Hanevik K. Human Mucosal IgA Immune Responses against Enterotoxigenic Escherichia coli. Pathogens 2020; 9:pathogens9090714. [PMID: 32872549 PMCID: PMC7558491 DOI: 10.3390/pathogens9090714] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/11/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022] Open
Abstract
Infection with enterotoxigenic Escherichia coli (ETEC) is a major contributor to diarrheal illness in children in low- and middle-income countries and travelers to these areas. There is an ongoing effort to develop vaccines against ETEC, and the most reliable immune correlate of protection against ETEC is considered to be the small intestinal secretory IgA response that targets ETEC-specific virulence factors. Since isolating IgA from small intestinal mucosa is technically and ethically challenging, requiring the use of invasive medical procedures, several other indirect methods are used as a proxy for gauging the small intestinal IgA responses. In this review, we summarize the literature reporting on anti-ETEC human IgA responses observed in blood, activated lymphocyte assayss, intestinal lavage/duodenal aspirates, and saliva from human volunteers being experimentally infected with ETEC. We describe the IgA response kinetics and responder ratios against classical and noncanonical ETEC antigens in the different sample types and discuss the implications that the results may have on vaccine development and testing.
Collapse
Affiliation(s)
- Saman Riaz
- Department of Clinical Science, University of Bergen, Jonas Lies veg 87, N-5021 Bergen, Norway;
- Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway
| | - Hans Steinsland
- Centre for Intervention Science in Maternal and Child Health, Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway;
- Department of Biomedicine, University of Bergen, 5020 Bergen, Norway
| | - Kurt Hanevik
- Department of Clinical Science, University of Bergen, Jonas Lies veg 87, N-5021 Bergen, Norway;
- Norwegian National Advisory Unit on Tropical Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Correspondence: ; Tel.: +47-5597-5000; Fax: +47-5597-2950
| |
Collapse
|
26
|
Bekeredjian-Ding I, Van Molle W, Baay M, Neels P. Human challenge trial workshop: Focus on quality requirements for challenge agents, Langen, Germany, October 22, 2019. Biologicals 2020; 66:53-61. [PMID: 32389512 DOI: 10.1016/j.biologicals.2020.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 01/01/2023] Open
Abstract
Controlled human infection models can be helpful to study pathogenesis and immune responses as a basis for the development of vaccines. In controlled human infection models, human challenge agents are used to infect healthy volunteers, therefore, ethical considerations include that the exposure studies need to be safe and results should be meaningful, e.g. contribute to a better cure. Both in the US and in Europe, the level of Good Manufacturing Practice required is related to the phase of the study ('sliding scale Good Manufacturing Practice'), and, hence, is much more open to speedy drug development than anticipated. Recommendations included: the development of guidelines for human challenge agents; a focus on strain selection, in particular with regard to strain infectivity, stability and purity; the use of whole genome sequencing; a reference repository of challenge agents, the need for early exchange with regulators to ensure acceptability of strain selection and manufacturing for later drug development; sharing of models and challenge agents.
Collapse
Affiliation(s)
- Isabelle Bekeredjian-Ding
- Paul-Ehrlich-Institut (PEI), Langen, Germany; Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany.
| | | | - Marc Baay
- P95 Epidemiology & Pharmacovigilance, Leuven, Belgium.
| | - Pieter Neels
- International Alliance for Biological Standardization (IABS), Belgium.
| | | |
Collapse
|
27
|
Olson ND, Kumar MS, Li S, Braccia DJ, Hao S, Timp W, Salit ML, Stine OC, Bravo HC. A framework for assessing 16S rRNA marker-gene survey data analysis methods using mixtures. MICROBIOME 2020; 8:35. [PMID: 32169095 PMCID: PMC7071580 DOI: 10.1186/s40168-020-00812-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 02/25/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND There are a variety of bioinformatic pipelines and downstream analysis methods for analyzing 16S rRNA marker-gene surveys. However, appropriate assessment datasets and metrics are needed as there is limited guidance to decide between available analysis methods. Mixtures of environmental samples are useful for assessing analysis methods as one can evaluate methods based on calculated expected values using unmixed sample measurements and the mixture design. Previous studies have used mixtures of environmental samples to assess other sequencing methods such as RNAseq. But no studies have used mixtures of environmental to assess 16S rRNA sequencing. RESULTS We developed a framework for assessing 16S rRNA sequencing analysis methods which utilizes a novel two-sample titration mixture dataset and metrics to evaluate qualitative and quantitative characteristics of count tables. Our qualitative assessment evaluates feature presence/absence exploiting features only present in unmixed samples or titrations by testing if random sampling can account for their observed relative abundance. Our quantitative assessment evaluates feature relative and differential abundance by comparing observed and expected values. We demonstrated the framework by evaluating count tables generated with three commonly used bioinformatic pipelines: (i) DADA2 a sequence inference method, (ii) Mothur a de novo clustering method, and (iii) QIIME an open-reference clustering method. The qualitative assessment results indicated that the majority of Mothur and QIIME features only present in unmixed samples or titrations were accounted for by random sampling alone, but this was not the case for DADA2 features. Combined with count table sparsity (proportion of zero-valued cells in a count table), these results indicate DADA2 has a higher false-negative rate whereas Mothur and QIIME have higher false-positive rates. The quantitative assessment results indicated the observed relative abundance and differential abundance values were consistent with expected values for all three pipelines. CONCLUSIONS We developed a novel framework for assessing 16S rRNA marker-gene survey methods and demonstrated the framework by evaluating count tables generated with three bioinformatic pipelines. This framework is a valuable community resource for assessing 16S rRNA marker-gene survey bioinformatic methods and will help scientists identify appropriate analysis methods for their marker-gene surveys.
Collapse
Affiliation(s)
- Nathan D. Olson
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, 100 Bureau Dr., Gaithersburg, 20899 MD USA
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, 8314 Paint Branch Dr., College Park, 20742 MD USA
- University of Maryland Institute of Advanced Computer Studies, University of Maryland, College Park, 8223 Paint Branch Dr., College Park, 20742 MD USA
| | - M. Senthil Kumar
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, 8314 Paint Branch Dr., College Park, 20742 MD USA
- University of Maryland Institute of Advanced Computer Studies, University of Maryland, College Park, 8223 Paint Branch Dr., College Park, 20742 MD USA
| | - Shan Li
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, 660 W. Redwood St., Baltimore, 21201 MD USA
| | - Domenick J. Braccia
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, 8314 Paint Branch Dr., College Park, 20742 MD USA
- University of Maryland Institute of Advanced Computer Studies, University of Maryland, College Park, 8223 Paint Branch Dr., College Park, 20742 MD USA
| | - Stephanie Hao
- Department of Biomedical Engineering, Johns Hopkins University, 720 Rutland Ave., Baltimore, 21205 MD USA
| | - Winston Timp
- Department of Biomedical Engineering, Johns Hopkins University, 720 Rutland Ave., Baltimore, 21205 MD USA
| | - Marc L. Salit
- Joint Initiative for Metrology in Biology, 443 Via Ortega, Stanford, 94305 CA USA
| | - O. Colin Stine
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, 660 W. Redwood St., Baltimore, 21201 MD USA
| | - Hector Corrada Bravo
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, 8314 Paint Branch Dr., College Park, 20742 MD USA
- University of Maryland Institute of Advanced Computer Studies, University of Maryland, College Park, 8223 Paint Branch Dr., College Park, 20742 MD USA
- Department of Computer Science, University of Maryland, College Park, 8223 Paint Branch Dr., College Park, 20742 MD USA
| |
Collapse
|
28
|
Savarino SJ, McKenzie R, Tribble DR, Porter CK, O'Dowd A, Sincock SA, Poole ST, DeNearing B, Woods CM, Kim H, Grahek SL, Brinkley C, Crabb JH, Bourgeois AL. Hyperimmune Bovine Colostral Anti-CS17 Antibodies Protect Against Enterotoxigenic Escherichia coli Diarrhea in a Randomized, Doubled-Blind, Placebo-Controlled Human Infection Model. J Infect Dis 2020; 220:505-513. [PMID: 30897198 DOI: 10.1093/infdis/jiz135] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 03/20/2019] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Enterotoxigenic Escherichia coli (ETEC) commonly cause diarrhea in children living in developing countries and in travelers to those regions. ETEC are characterized by colonization factors (CFs) that mediate intestinal adherence. We assessed if bovine colostral IgG (bIgG) antibodies against a CF, CS17, or antibodies against CsbD, the minor tip subunit of CS17, would protect subjects against diarrhea following challenge with a CS17-expressing ETEC strain. METHODS Adult subjects were randomized (1:1:1) to receive oral bIgG against CS17, CsbD, or placebo. Two days prior to challenge, subjects began dosing 3 times daily with the bIgG products (or placebo). On day 3, subjects ingested 5 × 109 cfu ETEC strain LSN03-016011/A in buffer. Subjects were assessed for diarrhea for 120 hours postchallenge. RESULTS A total of 36 subjects began oral prophylaxis and 35 were challenged with ETEC. While 50.0% of the placebo recipients had watery diarrhea, none of the subjects receiving anti-CS17 had diarrhea (P = .01). In contrast, diarrhea rates between placebo and anti-CsbD recipients (41.7%) were comparable (P = 1.0). CONCLUSIONS This is the first study to demonstrate anti-CS17 antibodies provide significant protection against ETEC expressing CS17. More research is needed to better understand why anti-CsbD was not comparably efficacious. Clinical Trials Registration. NCT00524004.
Collapse
Affiliation(s)
| | - Robin McKenzie
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.,Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | - Chad K Porter
- Naval Medical Research Center, Silver Spring, Maryland
| | | | | | | | - Barbara DeNearing
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | | - Hye Kim
- Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Shannon L Grahek
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Carl Brinkley
- Walter Reed Army Institute of Research, Silver Spring, Maryland
| | | | - A Louis Bourgeois
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
29
|
A new human challenge model for testing heat-stable toxin-based vaccine candidates for enterotoxigenic Escherichia coli diarrhea - dose optimization, clinical outcomes, and CD4+ T cell responses. PLoS Negl Trop Dis 2019; 13:e0007823. [PMID: 31665141 PMCID: PMC6844497 DOI: 10.1371/journal.pntd.0007823] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 11/11/2019] [Accepted: 10/02/2019] [Indexed: 12/17/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) are a common cause of diarrheal illness in young children and travelers. There is yet no licensed broadly protective vaccine against ETEC. One promising vaccine development strategy is to target strains expressing the heat-stable toxin (ST), particularly the human ST (STh), since infections with these strains are among the leading causes of diarrhea in children in low-and-middle income countries. A human challenge model based on an STh-only ETEC strain will be useful to evaluate the protective efficacy of new ST-based vaccine candidates. To develop this model, we experimentally infected 21 healthy adult volunteers with the epidemiologically relevant STh-only ETEC strain TW10722, identified a suitable dose, assessed safety, and characterized clinical outcomes and immune responses caused by the infection. Doses of 1×1010 colony-forming units (CFU) of TW10722 gave a suitable attack risk of 67% for moderate or severe diarrhea and an overall diarrhea attack risk of 78%. Non-diarrheal symptoms were mostly mild or moderate, and there were no serious adverse events. During the first month after ingesting the challenge strain, we measured significant increases in both activated CD4+ T cells and levels of serum IgG and IgA antibodies targeting coli surface antigen 5 (CS5) and 6 (CS6), as well as the E. coli mucinase YghJ. The CS5-specific CD4+ T cell and antibody responses were still significantly elevated one year after experimental infection. In conclusion, we have developed a safe STh-only ETEC-based human challenge model which can be efficiently used in Phase 2B trials to evaluate the protective efficacy of new ST-based vaccine candidates. Enterotoxigenic Escherichia coli (ETEC) is a common cause of diarrheal illness in young children living in low- and middle-income countries and in travelers to these countries. Several ETEC vaccine candidates are currently being developed, but so far, no broadly protective vaccines have been licensed. Since most moderate and severe ETEC diarrheal episodes are caused by strains that express the heat-stable enterotoxin (ST), ST represents a promising vaccine target. Here we present a human challenge model that can be used to estimate the protective efficacy of ST-based vaccine candidates in clinical vaccine trials. The model is based on the epidemiologically relevant ST-only ETEC strain TW10722, which we show is safe to ingest by volunteers and readily induce diarrhea.
Collapse
|
30
|
Chakraborty S, Randall A, Vickers TJ, Molina D, Harro CD, DeNearing B, Brubaker J, Sack DA, Bourgeois AL, Felgner PL, Liang X, Mani S, Wenzel H, Townsend RR, Gilmore PE, Darsley MJ, Rasko DA, Fleckenstein JM. Interrogation of a live-attenuated enterotoxigenic Escherichia coli vaccine highlights features unique to wild-type infection. NPJ Vaccines 2019; 4:37. [PMID: 31482013 PMCID: PMC6713706 DOI: 10.1038/s41541-019-0131-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 08/01/2019] [Indexed: 01/01/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) infections are a common cause of severe diarrheal illness in low- and middle-income countries. The live-attenuated ACE527 ETEC vaccine, adjuvanted with double mutant heat-labile toxin (dmLT), affords clear but partial protection against ETEC challenge in human volunteers. Comparatively, initial wild-type ETEC challenge completely protects against severe diarrhea on homologous re-challenge. To investigate determinants of protection, vaccine antigen content was compared to wild-type ETEC, and proteome microarrays were used to assess immune responses following vaccination and ETEC challenge. Although molecular interrogation of the vaccine confirmed expression of targeted canonical antigens, relative to wild-type ETEC, vaccine strains were deficient in production of flagellar antigens, immotile, and lacked production of the EtpA adhesin. Similarly, vaccination ± dmLT elicited responses to targeted canonical antigens, but relative to wild-type challenge, vaccine responses to some potentially protective non-canonical antigens including EtpA and the YghJ metalloprotease were diminished or absent. These studies highlight important differences in vaccine and wild-type ETEC antigen content and call attention to distinct immunologic signatures that could inform investigation of correlates of protection, and guide vaccine antigen selection for these pathogens of global importance.
Collapse
Affiliation(s)
| | | | - Tim J. Vickers
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, MO USA
| | - Doug Molina
- Antigen Discovery, Inc. (ADI), Irvine, CA USA
| | - Clayton D. Harro
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | | | - Jessica Brubaker
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | - David A. Sack
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | | | - Philip L. Felgner
- Antigen Discovery, Inc. (ADI), Irvine, CA USA
- Vaccine R & D Center, University of California, Irvine, Irvine, CA USA
| | | | - Sachin Mani
- Enteric Vaccine Initiative, PATH, Washington DC, USA
| | | | - R. Reid Townsend
- Department of Medicine, Divsion of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, USA
| | - Petra E. Gilmore
- Department of Medicine, Divsion of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, USA
| | | | - David A. Rasko
- The Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD USA
| | - James M. Fleckenstein
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, MO USA
- Medicine Service, John Cochran VA Medical Center, St. Louis, MO USA
| |
Collapse
|
31
|
Todnem Sakkestad S, Steinsland H, Skrede S, Kleppa E, Lillebø K, Sævik M, Søyland H, Rykkje Heien A, Gjerde Tellevik M, Barry EM, Sommerfelt H, Hanevik K. Experimental Infection of Human Volunteers with the Heat-Stable Enterotoxin-Producing Enterotoxigenic Escherichia coli Strain TW11681. Pathogens 2019; 8:pathogens8020084. [PMID: 31234485 PMCID: PMC6630672 DOI: 10.3390/pathogens8020084] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/17/2019] [Accepted: 06/20/2019] [Indexed: 12/03/2022] Open
Abstract
Infection with enterotoxigenic Escherichia coli (ETEC) producing the heat-stable enterotoxin (ST) is one of the most important causes of childhood diarrhoea in low- and middle-income countries. Here, we undertook a controlled human infection model (CHIM) study to investigate whether ST-producing ETEC strain TW11681 would be suitable for testing the protective efficacy of new ST-based vaccine candidates in vaccine challenge models. In groups of three, nine volunteers ingested 1 × 106, 1 × 107, or 1 × 108 colony-forming units (CFU) of TW11681. Flow cytometry-based assays were used to measure CD4+ T cell responses and antibody levels targeting virulence factors expressed by the strain. We found that infection with TW11681 elicited few and mild symptoms, including mild diarrhoea in two volunteers, both of whom ingested 1 × 106 CFU. Averaged across all volunteers, the CD4+ T cell responses specific for E. coli YghJ mucinase peaked 10 days after infection (3.2-fold (p = 0.016)), while the CD4+ T cell responses specific for Colonization Factor Antigen I (CFA/I) major fimbrial subunit (CfaB) peaked after 28 days (3.6-fold (p = 0.063)). The serum CfaB-specific anti-IgA and anti-IgG/IgM levels were significantly increased and peaked 3 months after infection. Both remained elevated for the duration of the 12-month follow-up. The corresponding anti-YghJ serological response was strongest after 10 days, although a significant increase was seen only for IgA levels (3.2-fold (p = 0.008)). In conclusion, due to its low diarrhoea attack risk, TW11681 is probably not suitable for testing the efficacy of new vaccines in human challenge studies at doses 1 × 106 to 1 × 108. However, the strain may still be useful in CHIMs for studying ETEC host-pathogen interactions.
Collapse
Affiliation(s)
- Sunniva Todnem Sakkestad
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Postbox 7804, 5020 Bergen, Norway; (S.T.S.); steinar.skrede@helse-bergen-no (S.S.)
| | - Hans Steinsland
- Centre for International Health, Department of Global Public Health and Primary Care, Faculty of Medicine, University of Bergen, Postbox 7894, 5020 Bergen, Norway;
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Postbox 7804, 5020 Bergen, Norway
| | - Steinar Skrede
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Postbox 7804, 5020 Bergen, Norway; (S.T.S.); steinar.skrede@helse-bergen-no (S.S.)
- Division for Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (K.L.); (M.S.); (H.S.); (A.R.H.)
| | - Elisabeth Kleppa
- Department of Microbiology, Haukeland University Hospital, 5021 Bergen, Norway;
| | - Kristine Lillebø
- Division for Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (K.L.); (M.S.); (H.S.); (A.R.H.)
| | - Marianne Sævik
- Division for Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (K.L.); (M.S.); (H.S.); (A.R.H.)
| | - Hanne Søyland
- Division for Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (K.L.); (M.S.); (H.S.); (A.R.H.)
| | - Astrid Rykkje Heien
- Division for Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (K.L.); (M.S.); (H.S.); (A.R.H.)
| | - Marit Gjerde Tellevik
- Norwegian National Advisory Unit on Tropical Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway;
| | - Eileen M. Barry
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Halvor Sommerfelt
- Centre for Intervention Science in Material and Child Health (CISMAC), Department of Global Public Health and Primary Care, Faculty of Medicine, University of Bergen, Postbox 7804, 5020 Bergen, Norway;
- Norwegian Institute of Public Health, 222 Oslo Postbox, Norway
| | - Kurt Hanevik
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Postbox 7804, 5020 Bergen, Norway; (S.T.S.); steinar.skrede@helse-bergen-no (S.S.)
- Norwegian National Advisory Unit on Tropical Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway;
- Correspondence: ; Tel.: +47-55-97-30-79
| |
Collapse
|
32
|
Clinical endpoints for efficacy studies. Vaccine 2019; 37:4814-4822. [PMID: 30981626 DOI: 10.1016/j.vaccine.2019.03.051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/19/2019] [Accepted: 03/21/2019] [Indexed: 12/21/2022]
Abstract
Well-established, validated and clinically meaningful primary and secondary endpoints are critical in advancing vaccines through proof of principal studies, licensure and pre-qualification. To that end, the field of vaccine development for Shigella, enterotoxigenic Escherichia coli (ETEC) as well as other enteric pathogens would benefit greatly from a focused review of clinical endpoints and the use of common endpoints across the field to enable study-to-study comparisons as well as comparative assessments between vaccine candidates. A workshop was conducted to review clinical endpoints from controlled human challenge studies, field studies in naïve adult travelers and pediatric studies in low-middle income countries and to develop a consensus on clinical endpoints for future vaccine trials. Following sequential presentations on different study designs (CHIM, travelers' efficacy and pediatric efficacy), workshop participants broke into three simultaneous workgroups focused on those study designs to discuss a number of topics key to clinical endpoints specific to each study design. Previously utilized endpoints were reviewed with an eye towards potentially novel endpoints for future studies and consideration of the disease parameters and spectrum of disease targeted for prevention. The strength of support among workshop participants for the use of various endpoints is summarized as are recommendations for additional endpoints to be considered in future studies. It is anticipated that this report will facilitate endpoint determination in future efficacy trials of vaccine candidates.
Collapse
|
33
|
Abstract
Zika virus (ZIKV), a previously little known arbovirus, caused an unprecedented outbreak in Latin America and the Caribbean throughout 2015 and 2016. The virus has been associated with the congenital Zika syndrome (CZS), which can occur with maternal ZIKV infection during any trimester and can result from asymptomatic infection. There is concern that even low levels of viremia can result in CZS, meaning an effective vaccine will need to induce very high levels of protection. Controlled human infection models (CHIMs), in which subjects are infected with a pathogen of interest, have been used to down-select vaccine candidates and have provided efficacy data in support of vaccine licensure.A ZIKV CHIM could be instrumental in determining which of the many ZIKV vaccine candidates provides the highest degree of protection and should be advanced in clinical development. The development of a ZIKV CHIM is not without challenges. The ZIKV, unlike other flaviviruses, is sexually and mosquito-transmitted, and an increase in the incidence of Guillain-Barré syndrome was reported in some countries during the ZIKV outbreak. These obstacles can be overcome with thoughtful study design to ensure maximal risk mitigation. If successful, a ZIKV CHIM could de-risk and accelerate ZIKV vaccine development.
Collapse
Affiliation(s)
- Anna P Durbin
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Stephen S Whitehead
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
34
|
Levine MM, Barry EM, Chen WH. A roadmap for enterotoxigenic Escherichia coli vaccine development based on volunteer challenge studies. Hum Vaccin Immunother 2019; 15:1357-1378. [PMID: 30724648 PMCID: PMC6663128 DOI: 10.1080/21645515.2019.1578922] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major cause of travelers’ diarrhea and of diarrhea among young children in developing countries. Experimental challenge studies in adult volunteers have played a pivotal role in establishing ETEC as an enteric pathogen, elucidating its pathogenesis by identifying specific virulence attributes, characterizing the human immune response to clinical and sub-clinical ETEC infection and assessing preliminarily the clinical acceptability, immunogenicity and efficacy of prototype ETEC vaccines. This review provides a historical perspective of experimental challenge studies with ETEC. It summarizes pioneering early studies carried out by investigators at the University of Maryland School of Medicine to show how those studies provided key information that influenced the directions taken by many research groups to develop vaccines to prevent ETEC. In addition, key experimental challenge studies undertaken at other institutions will also be cited.
Collapse
Affiliation(s)
- Myron M Levine
- a Center for Vaccine Development and Global Health , University of Maryland School of Medicine , Baltimore , MD , USA
| | - Eileen M Barry
- a Center for Vaccine Development and Global Health , University of Maryland School of Medicine , Baltimore , MD , USA
| | - Wilbur H Chen
- a Center for Vaccine Development and Global Health , University of Maryland School of Medicine , Baltimore , MD , USA
| |
Collapse
|
35
|
Harro C, Louis Bourgeois A, Sack D, Walker R, DeNearing B, Brubaker J, Maier N, Fix A, Dally L, Chakraborty S, Clements JD, Saunders I, Darsley MJ. Live attenuated enterotoxigenic Escherichia coli (ETEC) vaccine with dmLT adjuvant protects human volunteers against virulent experimental ETEC challenge. Vaccine 2019; 37:1978-1986. [PMID: 30797634 PMCID: PMC6434318 DOI: 10.1016/j.vaccine.2019.02.025] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/12/2019] [Accepted: 02/14/2019] [Indexed: 01/10/2023]
Abstract
Background There is no licensed vaccine against enterotoxigenic Escherichia coli (ETEC), a major cause of diarrhea-associated morbidity and mortality among infants and children in low-income countries and travelers. The results of this vaccination/challenge study demonstrate strong protection by an attenuated ETEC vaccine candidate, ACE527, when co-administered with a mucosal adjuvant, the double-mutant heat-labile toxin (dmLT) of ETEC. Methods Sixty healthy adults participated in a randomized, placebo-controlled, double-blind study with three doses of lyophilized ACE527 (∼3 × 109 of each strain per dose) administered orally with or without dmLT adjuvant (25 µg/dose). Six months later, 36 of these volunteers and a control group of 21 unvaccinated volunteers were challenged with virulent ETEC strain H10407. The primary outcome was severe diarrhea, defined as passing >800 g of unformed stools during the inpatient period following challenge. Findings The vaccine was well tolerated and induced robust immune responses to key antigens. The protective efficacy (PE) against the primary outcome of severe diarrhea was 65.9% (95% confidence interval [CI] 5.4–87.7, p = 0.003). Among subjects receiving the adjuvanted vaccine, the attack rate of severe diarrhea was 23.1, while in unimmunized controls it was 67.7%. The PE against diarrhea of any severity was 58.5% (95% CI 3.8– 82.1, p = 0.016). There was a strong inverse correlation between shedding of the vaccine strain after either of the first two doses and absence of severe diarrhea upon challenge (RR = 0.29, 95% CI 0.08–1.05, p = 0.041). Challenge strain shedding was 10-fold lower in those receiving the adjuvant than in those receiving vaccine alone. The unadjuvanted vaccine was not protective (PE = 23.1%). Interpretation The results of this study support further development of ACE527 + dmLT as a vaccine for children in endemic countries and travelers. This is the first clinical demonstration that dmLT can contribute significantly to vaccine efficacy and may warrant testing with other oral vaccines. (ClinicalTrials.gov registration: NCT01739231).
Collapse
Affiliation(s)
- Clayton Harro
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - David Sack
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Barbara DeNearing
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jessica Brubaker
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | | | - Len Dally
- The Emmes Corporation, Rockville, MD, USA
| | - Subhra Chakraborty
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | | | | |
Collapse
|
36
|
Osowicki J, Azzopardi KI, McIntyre L, Rivera-Hernandez T, Ong CLY, Baker C, Gillen CM, Walker MJ, Smeesters PR, Davies MR, Steer AC. A Controlled Human Infection Model of Group A Streptococcus Pharyngitis: Which Strain and Why? mSphere 2019; 4:e00647-18. [PMID: 30760615 PMCID: PMC6374595 DOI: 10.1128/msphere.00647-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 01/16/2019] [Indexed: 01/23/2023] Open
Abstract
Group A Streptococcus (GAS) is a major cause of global infection-related morbidity and mortality. A modern controlled human infection model (CHIM) of GAS pharyngitis can accelerate vaccine development and pathogenesis research. A robust rationale for strain selection is central to meeting ethical, scientific, and regulatory requirements. Multifaceted characterization studies were done to compare a preferred candidate emm75 (M75) GAS strain to three other strains: an alternative candidate emm12 (M12) strain, an M1 strain used in 1970s pharyngitis CHIM studies (SS-496), and a representative (5448) of the globally disseminated M1T1 clone. A range of approaches were used to explore strain growth, adherence, invasion, delivery characteristics, short- and long-term viability, phylogeny, virulence factors, vaccine antigens, resistance to killing by human neutrophils, and lethality in a murine invasive model. The strains grew reliably in a medium without animal-derived components, were consistently transferred using a swab method simulating the CHIM protocol, remained viable at -80°C, and carried genes for most candidate vaccine antigens. Considering GAS molecular epidemiology, virulence factors, in vitro assays, and results from the murine model, the contemporary strains show a spectrum of virulence, with M75 appearing the least virulent and 5448 the most. The virulence profile of SS-496, used safely in 1970s CHIM studies, was similar to that of 5448 in the animal model and virulence gene carriage. The results of this multifaceted characterization confirm the M75 strain as an appropriate choice for initial deployment in the CHIM, with the aim of safely and successfully causing pharyngitis in healthy adult volunteers.IMPORTANCE GAS (Streptococcus pyogenes) is a leading global cause of infection-related morbidity and mortality. A modern CHIM of GAS pharyngitis could help to accelerate vaccine development and drive pathogenesis research. Challenge strain selection is critical to the safety and success of any CHIM and especially so for an organism such as GAS, with its wide strain diversity and potential to cause severe life-threatening acute infections (e.g., toxic shock syndrome and necrotizing fasciitis) and postinfectious complications (e.g., acute rheumatic fever, rheumatic heart disease, and acute poststreptococcal glomerulonephritis). In this paper, we outline the rationale for selecting an emm75 strain for initial use in a GAS pharyngitis CHIM in healthy adult volunteers, drawing on the findings of a broad characterization effort spanning molecular epidemiology, in vitro assays, whole-genome sequencing, and animal model studies.
Collapse
Affiliation(s)
- Joshua Osowicki
- Tropical Diseases, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Infectious Diseases Unit, Department of General Medicine, The Royal Children's Hospital Melbourne, Melbourne, Victoria, Australia
| | - Kristy I Azzopardi
- Tropical Diseases, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Liam McIntyre
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Tania Rivera-Hernandez
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Queensland, Australia
| | - Cheryl-Lynn Y Ong
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Queensland, Australia
| | - Ciara Baker
- Tropical Diseases, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Christine M Gillen
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Queensland, Australia
| | - Mark J Walker
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Queensland, Australia
| | - Pierre R Smeesters
- Tropical Diseases, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Paediatric Department, Academic Children Hospital Queen Fabiola, Université Libre de Bruxelles, Brussels, Belgium
- Molecular Bacteriology Laboratory, Université Libre de Bruxelles, Brussels, Belgium
| | - Mark R Davies
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Andrew C Steer
- Tropical Diseases, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Infectious Diseases Unit, Department of General Medicine, The Royal Children's Hospital Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
37
|
Hanevik K, Chen WH, Talaat KR, Porter C, Bourgeois L. The way forward for ETEC controlled human infection models (CHIMs). Vaccine 2019; 37:4794-4799. [PMID: 30709728 DOI: 10.1016/j.vaccine.2019.01.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/10/2018] [Accepted: 01/03/2019] [Indexed: 12/27/2022]
Abstract
In the absence of good animal models, Controlled Human Infection Models (CHIMs) are useful to assess efficacy of new vaccine candidates against Enterotoxic Escherichia coli (ETEC), as well as other preventive or therapeutic interventions. At the 2018 Vaccines Against Shigella and ETEC (VASE) conference, a workshop was held to further review and discuss new challenge model developments and key issues related to further model standardization. During the workshop, invited speakers briefly summarized for attendees recent developments and main agenda issues before workshop participants were divided into four groups for more focused discussions. The main issues discussed were: (1) whether there is a need for more ETEC strains to test a diversity of vaccine candidates, and if so, what criteria/qualities are desirable in strain selection; (2) how ETEC CHIMs could be more standardized to better support ETEC vaccine development; (3) how volunteer selection criteria and screening should be performed, and; (4) how an expanded sample collection schema and collaborative analysis plan may facilitate a more in-depth assessment of the role of antigen-specific humoral and cellular immune responses in ETEC infection, and provide better insights into ETEC pathogenesis and correlates of protection. The workshop concluded that additional challenge strains may need to be developed to better support new vaccines and therapeutics that are advancing in the development pipeline. In this regard, the need for a well characterized ST-only expressing ETEC strain was highlighted as a priority given that promising new heat stable toxoid based vaccine candidates are on the horizon. In addition, further standardization of the ETEC CHIMs was strongly encouraged, noting that it may not be realistic to standardize across all strains. Also, intensified volunteer screening may result in higher attack rates, although more stringent eligibility criteria may contribute to a more limited application of the model and diminish its representativeness. Finally, a sampling schedule and priority list for minimum set of samples was also proposed. Future workshops could be held to further refine standards for ETEC CHIMS and to facilitate more collaborative work on stored sample sets from previous and future ETEC CHIMs to maximize the contribution of these trials to our understanding of ETEC pathogenesis and our development of better prevention and control measures for this important pathogen.
Collapse
Affiliation(s)
- Kurt Hanevik
- Norwegian National Advisory Unit on Tropical Infectious Diseases, Department of Medicine, Haukeland University Hospital, Bergen, Norway; Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway.
| | - Wilbur H Chen
- University of Maryland School of Medicine, Center for Vaccine Development, Baltimore, MD, USA
| | - Kawsar R Talaat
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore MD, USA
| | - Chad Porter
- Naval Medical Research Center, Forest Glen, MD, USA
| | | |
Collapse
|
38
|
Fleckenstein JM. Providing Structure to Enterotoxigenic Escherichia coli Vaccine Development. J Infect Dis 2018; 216:1-3. [PMID: 28541516 DOI: 10.1093/infdis/jix146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 03/17/2017] [Indexed: 01/09/2023] Open
Affiliation(s)
- James M Fleckenstein
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine.,Medicine Service, Veterans Affairs Medical Center, St. Louis, Missouri
| |
Collapse
|
39
|
Chakraborty S, Randall A, Vickers TJ, Molina D, Harro CD, DeNearing B, Brubaker J, Sack DA, Bourgeois AL, Felgner PL, Liang X, Mani S, Wenzel H, Townsend RR, Gilmore PE, Darsley MJ, Rasko DA, Fleckenstein JM. Human Experimental Challenge With Enterotoxigenic Escherichia coli Elicits Immune Responses to Canonical and Novel Antigens Relevant to Vaccine Development. J Infect Dis 2018; 218:1436-1446. [PMID: 29800314 PMCID: PMC6151082 DOI: 10.1093/infdis/jiy312] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 05/22/2018] [Indexed: 11/12/2022] Open
Abstract
Background Enterotoxigenic Escherichia coli (ETEC) is a major cause of diarrheal illness in the developing world. Enterotoxigenic E coli vaccinology has been challenged by genetic diversity and heterogeneity of canonical antigens. Examination of the antigenic breadth of immune responses associated with protective immunity could afford new avenues for vaccine development. Methods Antibody lymphocyte supernatants (ALS) and sera from 20 naive human volunteers challenged with ETEC strain H10407 and from 10 volunteers rechallenged 4-6 weeks later with the same strain (9 of whom were completely protected on rechallenge) were tested against ETEC proteome microarrays containing 957 antigens. Results Enterotoxigenic E coli challenge stimulated robust serum and mucosal (ALS) responses to canonical vaccine antigens (CFA/I, and the B subunit of LT) as well as a small number of antigens not presently targeted in ETEC vaccines. These included pathovar-specific secreted proteins (EtpA, EatA) as well as highly conserved E coli antigens including YghJ, flagellin, and pertactin-like autotransporter proteins, all of which have previously afforded protection against ETEC infection in preclinical studies. Conclusions Taken together, studies reported here suggest that immune responses after ETEC infection involve traditional vaccine targets as well as a select number of more recently identified protein antigens that could offer additional avenues for vaccine development for these pathogens.
Collapse
Affiliation(s)
| | - Arlo Randall
- Antigen Discovery, Inc. (ADI), Irvine, California
| | - Tim J Vickers
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri
| | - Doug Molina
- Antigen Discovery, Inc. (ADI), Irvine, California
| | - Clayton D Harro
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Barbara DeNearing
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Jessica Brubaker
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - David A Sack
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | | | | - Xiaowu Liang
- Antigen Discovery, Inc. (ADI), Irvine, California
| | | | | | - R Reid Townsend
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine
| | - Petra E Gilmore
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine
| | | | - David A Rasko
- The Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - James M Fleckenstein
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri
- Medicine Service, John Cochran VA Medical Center, St. Louis, Missouri
| |
Collapse
|
40
|
Crofts AA, Giovanetti SM, Rubin EJ, Poly FM, Gutiérrez RL, Talaat KR, Porter CK, Riddle MS, DeNearing B, Brubaker J, Maciel M, Alcala AN, Chakraborty S, Prouty MG, Savarino SJ, Davies BW, Trent MS. Enterotoxigenic E. coli virulence gene regulation in human infections. Proc Natl Acad Sci U S A 2018; 115:E8968-E8976. [PMID: 30126994 PMCID: PMC6156659 DOI: 10.1073/pnas.1808982115] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a global diarrheal pathogen that utilizes adhesins and secreted enterotoxins to cause disease in mammalian hosts. Decades of research on virulence factor regulation in ETEC has revealed a variety of environmental factors that influence gene expression, including bile, pH, bicarbonate, osmolarity, and glucose. However, other hallmarks of the intestinal tract, such as low oxygen availability, have not been examined. Further, determining how ETEC integrates these signals in the complex host environment is challenging. To address this, we characterized ETEC's response to the human host using samples from a controlled human infection model. We found ETEC senses environmental oxygen to globally influence virulence factor expression via the oxygen-sensitive transcriptional regulator fumarate and nitrate reduction (FNR) regulator. In vitro anaerobic growth replicates the in vivo virulence factor expression profile, and deletion of fnr in ETEC strain H10407 results in a significant increase in expression of all classical virulence factors, including the colonization factor antigen I (CFA/I) adhesin operon and both heat-stable and heat-labile enterotoxins. These data depict a model of ETEC infection where FNR activity can globally influence virulence gene expression, and therefore proximity to the oxygenated zone bordering intestinal epithelial cells likely influences ETEC virulence gene expression in vivo. Outside of the host, ETEC biofilms are associated with seasonal ETEC epidemics, and we find FNR is a regulator of biofilm production. Together these data suggest FNR-dependent oxygen sensing in ETEC has implications for human infection inside and outside of the host.
Collapse
Affiliation(s)
- Alexander A Crofts
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
| | - Simone M Giovanetti
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
| | - Erica J Rubin
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
| | - Frédéric M Poly
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD 20910
| | - Ramiro L Gutiérrez
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD 20910
| | - Kawsar R Talaat
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| | - Chad K Porter
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD 20910
| | - Mark S Riddle
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD 20910
| | - Barbara DeNearing
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| | - Jessica Brubaker
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| | - Milton Maciel
- Immunology, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Ashley N Alcala
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD 20910
| | - Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| | - Michael G Prouty
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD 20910
| | - Stephen J Savarino
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD 20910
| | - Bryan W Davies
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
| | - M Stephen Trent
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602;
- Center of Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
- Department of Microbiology, College of Arts and Sciences, University of Georgia, Athens, GA 30602
| |
Collapse
|
41
|
Kumar P, Kuhlmann FM, Chakraborty S, Bourgeois AL, Foulke-Abel J, Tumala B, Vickers TJ, Sack DA, DeNearing B, Harro CD, Wright WS, Gildersleeve JC, Ciorba MA, Santhanam S, Porter CK, Gutierrez RL, Prouty MG, Riddle MS, Polino A, Sheikh A, Donowitz M, Fleckenstein JM. Enterotoxigenic Escherichia coli-blood group A interactions intensify diarrheal severity. J Clin Invest 2018; 128:3298-3311. [PMID: 29771685 DOI: 10.1172/jci97659] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 05/03/2018] [Indexed: 12/27/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) infections are highly prevalent in developing countries, where clinical presentations range from asymptomatic colonization to severe cholera-like illness. The molecular basis for these varied presentations, which may involve strain-specific virulence features as well as host factors, has not been elucidated. We demonstrate that, when challenged with ETEC strain H10407, originally isolated from a case of cholera-like illness, blood group A human volunteers developed severe diarrhea more frequently than individuals from other blood groups. Interestingly, a diverse population of ETEC strains, including H10407, secrete the EtpA adhesin molecule. As many bacterial adhesins also agglutinate red blood cells, we combined the use of glycan arrays, biolayer inferometry, and noncanonical amino acid labeling with hemagglutination studies to demonstrate that EtpA is a dominant ETEC blood group A-specific lectin/hemagglutinin. Importantly, we have also shown that EtpA interacts specifically with glycans expressed on intestinal epithelial cells from blood group A individuals and that EtpA-mediated bacterial-host interactions accelerate bacterial adhesion and effective delivery of both the heat-labile and heat-stable toxins of ETEC. Collectively, these data provide additional insight into the complex molecular basis of severe ETEC diarrheal illness that may inform rational design of vaccines to protect those at highest risk.
Collapse
Affiliation(s)
- Pardeep Kumar
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri, USA
| | - F Matthew Kuhlmann
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - A Louis Bourgeois
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jennifer Foulke-Abel
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Brunda Tumala
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tim J Vickers
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David A Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Barbara DeNearing
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Clayton D Harro
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - W Shea Wright
- Center for Cancer Research, Chemical Biology Laboratory, National Cancer Institute, Fredrick, Maryland, USA
| | - Jeffrey C Gildersleeve
- Center for Cancer Research, Chemical Biology Laboratory, National Cancer Institute, Fredrick, Maryland, USA
| | - Matthew A Ciorba
- Department of Medicine, Division of Gastroenterology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Srikanth Santhanam
- Department of Medicine, Division of Gastroenterology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Chad K Porter
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Ramiro L Gutierrez
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Michael G Prouty
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Mark S Riddle
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Alexander Polino
- Molecular Microbiology and Microbial Pathogenesis Program, Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alaullah Sheikh
- Molecular Microbiology and Microbial Pathogenesis Program, Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mark Donowitz
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - James M Fleckenstein
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri, USA.,Molecular Microbiology and Microbial Pathogenesis Program, Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, Missouri, USA.,Medicine Service, Veterans Affairs Medical Center, St. Louis, Missouri, USA
| |
Collapse
|
42
|
Chakraborty S, Harro C, DeNearing B, Brubaker J, Connor S, Maier N, Dally L, Flores J, Bourgeois AL, Walker R, Sack DA. Impact of lower challenge doses of enterotoxigenic Escherichia coli on clinical outcome, intestinal colonization and immune responses in adult volunteers. PLoS Negl Trop Dis 2018; 12:e0006442. [PMID: 29702652 PMCID: PMC5942845 DOI: 10.1371/journal.pntd.0006442] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 05/09/2018] [Accepted: 04/10/2018] [Indexed: 11/18/2022] Open
Abstract
A reliable and effective human challenge model is needed to help down-select the most promising ETEC vaccines currently under development. Such a model would need to reliably induce diarrhea in a high proportion of volunteers using the lowest possible inoculum to maximize safety and sensitivity. Previously we validated a challenge model that utilized a dose of 2x107 CFU of ETEC strain H10407 (LT+, ST+, CFA/I+ and O78+) to induce attack rates for moderate to severe diarrhea (MSD) of ~60-70%. Here we detail efforts to further refine the model in an attempt to determine if a lower challenge dose of H10407 can be used. Thirty subjects were randomized 1:1 to receive an oral administration of H10407 at doses of 106 or 105 CFU in bicarbonate buffer. After challenge, subjects were monitored for signs and symptoms of enteric illness and stool samples were collected to detect shedding of the challenge strain. Systemic and mucosal immune responses were measured using serum, antibody in lymphocyte supernatant and fecal samples. The attack rate was 13.3% (2/15) and 26.7% (4/15) for MSD in the 105 and 106 groups, respectively. Four MSD cases met criteria for early antibiotic treatment. All subjects but one shed the challenge strain in fecal samples. The frequency and magnitude of anti-LT toxin, CFA/I and LPS O78 immune responses were antigen, dose, severity of diarrhea and shedding levels dependent. Notably, although of lower magnitude, there were considerable immune responses in the subjects with no diarrhea. This may indicate that immune responses to asymptomatic infections of ETEC in children in the endemic countries may contribute to protection. Based on this and our prior studies, we conclude that a dose of 2x107 H10407 remains the lowest practical dose for use in future volunteer studies evaluating candidate vaccines and other preventive or therapeutic ETEC interventions. TRIAL REGISTRATION ClinicalTrials.gov NCT00844493.
Collapse
Affiliation(s)
- Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- * E-mail:
| | - Clayton Harro
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Barbara DeNearing
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Jessica Brubaker
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Sean Connor
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | | | - Len Dally
- The EMMES Corporation, Rockville, Maryland, United States of America
| | | | - A. Louis Bourgeois
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- PATH, Washington, DC, United States of America
| | | | - David A. Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
43
|
Baay MFD, Richie TL, Neels P. Human challenge trials in vaccine development, Rockville, MD, USA, September 28-30, 2017. Biologicals 2018; 61:85-94. [PMID: 29573967 DOI: 10.1016/j.biologicals.2018.02.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 02/21/2018] [Indexed: 11/17/2022] Open
Abstract
The International Alliance for Biological Standardization organized the second workshop on human challenge trials (HCT) in Rockville, MD, in September 2017. The objective of this meeting was to examine the use of HCT, in response to the continuing human suffering caused by infectious diseases, preventable by the development of new and improved vaccines. For this, the approach of HCT could be valuable, as HCT can provide key safety, tolerability, immunogenicity, and efficacy data, and can be used to study host-pathogen biology. HCT can generate these data with speed, efficiency and minimal expense, albeit not with the same level of robustness as clinical trials. Incorporated wisely into a clinical development plan, HCT can support optimization or down-selection of new vaccine candidates, assuring that only the worthiest candidates progress to field testing. HCT may also provide pivotal efficacy data in support of licensure, particularly when field efficacy studies are not feasible. Many aspects of HCT were discussed by the participants, including new and existing models, standardization and ethics. A consensus was achieved that HCT, if ethically justified and performed with careful attention to safety and informed consent, should be pursued to promote and accelerate vaccine development.
Collapse
Affiliation(s)
- Marc F D Baay
- P95 Pharmacovigilance and Epidemiology Services, Leuven, Belgium.
| | - Thomas L Richie
- Sanaria Institute for Global Health and Tropical Medicine, Rockville, MD, USA.
| | - Pieter Neels
- International Alliance for Biological Standardization, Lyon, France.
| | | |
Collapse
|
44
|
Rojas-Lopez M, Monterio R, Pizza M, Desvaux M, Rosini R. Intestinal Pathogenic Escherichia coli: Insights for Vaccine Development. Front Microbiol 2018; 9:440. [PMID: 29615989 PMCID: PMC5869917 DOI: 10.3389/fmicb.2018.00440] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 02/26/2018] [Indexed: 12/13/2022] Open
Abstract
Diarrheal diseases are one of the major causes of mortality among children under five years old and intestinal pathogenic Escherichia coli (InPEC) plays a role as one of the large causative groups of these infections worldwide. InPECs contribute significantly to the burden of intestinal diseases, which are a critical issue in low- and middle-income countries (Asia, Africa and Latin America). Intestinal pathotypes such as enteropathogenic E. coli (EPEC) and enterotoxigenic E. coli (ETEC) are mainly endemic in developing countries, while ETEC strains are the major cause of diarrhea in travelers to these countries. On the other hand, enterohemorrhagic E. coli (EHEC) are the cause of large outbreaks around the world, mainly affecting developed countries and responsible for not only diarrheal disease but also severe clinical complications like hemorrhagic colitis and hemolytic uremic syndrome (HUS). Overall, the emergence of antibiotic resistant strains, the annual cost increase in the health care system, the high incidence of traveler diarrhea and the increased number of HUS episodes have raised the need for effective preventive treatments. Although the use of antibiotics is still important in treating such infections, non-antibiotic strategies are either a crucial option to limit the increase in antibiotic resistant strains or absolutely necessary for diseases such as those caused by EHEC infections, for which antibiotic therapies are not recommended. Among non-antibiotic therapies, vaccine development is a strategy of choice but, to date, there is no effective licensed vaccine against InPEC infections. For several years, there has been a sustained effort to identify efficacious vaccine candidates able to reduce the burden of diarrheal disease. The aim of this review is to summarize recent milestones and insights in vaccine development against InPECs.
Collapse
Affiliation(s)
- Maricarmen Rojas-Lopez
- GSK, Siena, Italy.,Institut National de la Recherche Agronomique, Université Clermont Auvergne, UMR454 MEDiS, Clermont-Ferrand, France
| | - Ricardo Monterio
- Institut National de la Recherche Agronomique, Université Clermont Auvergne, UMR454 MEDiS, Clermont-Ferrand, France
| | | | - Mickaël Desvaux
- Institut National de la Recherche Agronomique, Université Clermont Auvergne, UMR454 MEDiS, Clermont-Ferrand, France
| | | |
Collapse
|
45
|
Begum YA, Rydberg HA, Thorell K, Kwak YK, Sun L, Joffré E, Qadri F, Sjöling Å. In Situ Analyses Directly in Diarrheal Stool Reveal Large Variations in Bacterial Load and Active Toxin Expression of Enterotoxigenic Escherichiacoli and Vibrio cholerae. mSphere 2018; 3:e00517-17. [PMID: 29404412 PMCID: PMC5784243 DOI: 10.1128/msphere.00517-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/27/2017] [Indexed: 11/20/2022] Open
Abstract
The bacterial pathogens enterotoxigenic Escherichia coli (ETEC) and Vibrio cholerae are major causes of diarrhea. ETEC causes diarrhea by production of the heat-labile toxin (LT) and heat-stable toxins (STh and STp), while V. cholerae produces cholera toxin (CT). In this study, we determined the occurrence and bacterial doses of the two pathogens and their respective toxin expression levels directly in liquid diarrheal stools of patients in Dhaka, Bangladesh. By quantitative culture and real-time quantitative PCR (qPCR) detection of the toxin genes, the two pathogens were found to coexist in several of the patients, at concentrations between 102 and 108 bacterial gene copies per ml. Even in culture-negative samples, gene copy numbers of 102 to 104 of either ETEC or V. cholerae toxin genes were detected by qPCR. RNA was extracted directly from stool, and gene expression levels, quantified by reverse transcriptase qPCR (RT-qPCR), of the genes encoding CT, LT, STh, and STp showed expression of toxin genes. Toxin enzyme-linked immunosorbent assay (ELISA) confirmed active toxin secretion directly in the liquid diarrhea. Analysis of ETEC isolates by multiplex PCR, dot blot analysis, and genome sequencing suggested that there are genetic ETEC profiles that are more commonly found as dominating single pathogens and others that are coinfectants with lower bacterial loads. The ETEC genomes, including assembled genomes of dominating ETEC isolates expressing LT/STh/CS5/CS6 and LT/CS7, are provided. In addition, this study highlights an emerging important ETEC strain expressing LT/STp and the novel colonization factor CS27b. These findings have implications for investigations of pathogenesis as well as for vaccine development. IMPORTANCE The cause of diarrheal disease is usually determined by screening for several microorganisms by various methods, and sole detection is used to assign the agent as the cause of disease. However, it has become increasingly clear that many infections are caused by coinfections with several pathogens and that the dose of the infecting pathogen is important. We quantified the absolute numbers of enterotoxigenic E. coli (ETEC) and Vibrio cholerae directly in diarrheal fluid. We noted several events where both pathogens were found but also a large dose dependency. In three samples, we found ETEC as the only pathogen sought for. These isolates belonged to globally distributed ETEC clones and were the dominating species in stool with active toxin expression. This suggests that certain superior virulent ETEC lineages are able to outcompete the gut microbiota and be the sole cause of disease and hence need to be specifically monitored.
Collapse
Affiliation(s)
- Yasmin Ara Begum
- International Centre for Diarrhoeal Disease Research, Bangladesh, Centre for Health and Population Research, Dhaka, Bangladesh
| | - Hanna A. Rydberg
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
| | - Kaisa Thorell
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
| | - Young-Keun Kwak
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
| | - Lei Sun
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
| | - Enrique Joffré
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
| | - Firdausi Qadri
- International Centre for Diarrhoeal Disease Research, Bangladesh, Centre for Health and Population Research, Dhaka, Bangladesh
| | - Åsa Sjöling
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
46
|
Savarino SJ, McKenzie R, Tribble DR, Porter CK, O'Dowd A, Cantrell JA, Sincock SA, Poole ST, DeNearing B, Woods CM, Kim H, Grahek SL, Brinkley C, Crabb JH, Bourgeois AL. Prophylactic Efficacy of Hyperimmune Bovine Colostral Antiadhesin Antibodies Against Enterotoxigenic Escherichia coli Diarrhea: A Randomized, Double-Blind, Placebo-Controlled, Phase 1 Trial. J Infect Dis 2017; 216:7-13. [PMID: 28541500 DOI: 10.1093/infdis/jix144] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/17/2017] [Indexed: 11/13/2022] Open
Abstract
Background Tip-localized adhesive proteins of bacterial fimbriae from diverse pathogens confer protection in animal models, but efficacy in humans has not been reported. Enterotoxigenic Escherichia coli (ETEC) commonly elaborate colonization factors comprising a minor tip adhesin and major stalk-forming subunit. We assessed the efficacy of antiadhesin bovine colostral IgG (bIgG) antibodies against ETEC challenge in volunteers. Methods Adults were randomly assigned (1:1:1) to take oral hyperimmune bIgG raised against CFA/I minor pilin subunit (CfaE) tip adhesin or colonization factor I (CFA/I) fimbraie (positive control) or placebo. Two days before challenge, volunteers began a thrice-daily, 7-day course of investigational product administered in sodium bicarbonate 15 minutes after each meal. On day 3, subjects drank 1 × 109 colony-forming units of colonization factor I (CFA/I)-ETEC strain H10407 with buffer. The primary efficacy endpoint was diarrhea within 120 hours of challenge. Results After enrollment and randomization, 31 volunteers received product, underwent ETEC challenge, and were included in the per protocol efficacy analysis. Nine of 11 placebos developed diarrhea, 7 experiencing moderate to severe disease. Protective efficacy of 63% (P = .03) and 88% (P = .002) was observed in the antiadhesin bIgG and positive control groups, respectively. Conclusions Oral administration of anti-CFA/I minor pilin subunit (CfaE) antibodies conferred significant protection against ETEC, providing the first clinical evidence that fimbrial tip adhesins function as protective antigens.
Collapse
Affiliation(s)
| | - Robin McKenzie
- Johns Hopkins Bloomberg School of Public Health.,Johns Hopkins School of Medicine, Baltimore
| | | | - Chad K Porter
- Naval Medical Research Center, Silver Spring, Maryland
| | | | | | | | | | | | | | - Hye Kim
- Johns Hopkins School of Medicine, Baltimore
| | | | - Carl Brinkley
- Walter Reed Army Institute of Research, Silver Spring, Maryland
| | | | | |
Collapse
|
47
|
Bron PA, Kleerebezem M, Brummer RJ, Cani PD, Mercenier A, MacDonald TT, Garcia-Ródenas CL, Wells JM. Can probiotics modulate human disease by impacting intestinal barrier function? Br J Nutr 2017; 117:93-107. [PMID: 28102115 PMCID: PMC5297585 DOI: 10.1017/s0007114516004037] [Citation(s) in RCA: 307] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 09/29/2016] [Accepted: 10/25/2016] [Indexed: 12/16/2022]
Abstract
Intestinal barrier integrity is a prerequisite for homeostasis of mucosal function, which is balanced to maximise absorptive capacity, while maintaining efficient defensive reactions against chemical and microbial challenges. Evidence is mounting that disruption of epithelial barrier integrity is one of the major aetiological factors associated with several gastrointestinal diseases, including infection by pathogens, obesity and diabetes, necrotising enterocolitis, irritable bowel syndrome and inflammatory bowel disease. The notion that specific probiotic bacterial strains can affect barrier integrity fuelled research in which in vitro cell lines, animal models and clinical trials are used to assess whether probiotics can revert the diseased state back to homeostasis and health. This review catalogues and categorises the lines of evidence available in literature for the role of probiotics in epithelial integrity and, consequently, their beneficial effect for the reduction of gastrointestinal disease symptoms.
Collapse
Affiliation(s)
- Peter A. Bron
- NIZO Food Research and BE-Basic Foundation, Kernhemseweg 2, 6718ZB Ede, The Netherlands
| | - Michiel Kleerebezem
- Wageningen University, Host Microbe Interactomics Group, De Elst 1, 6708WD Wageningen, The Netherlands
| | - Robert-Jan Brummer
- Faculty of Medicine and Health, Örebro University, Fakultetsgatan 1, SE-701 82 Örebro, Sweden
| | - Patrice D. Cani
- Metabolism and Nutrition Research Group, WELBIO – Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Université catholique de Louvain, Avenue E. Mounier, 73 B1.73.11, 1200 Brussels, Belgium
| | - Annick Mercenier
- Nestlé Research Center, Nutrition and Health Research, route du Jorat 57, 1000 Lausanne 26, Switzerland
| | - Thomas T. MacDonald
- Barts and The London school of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Clara L. Garcia-Ródenas
- Nestlé Research Center, Nutrition and Health Research, route du Jorat 57, 1000 Lausanne 26, Switzerland
| | - Jerry M. Wells
- Wageningen University, Host Microbe Interactomics Group, De Elst 1, 6708WD Wageningen, The Netherlands
| |
Collapse
|
48
|
Julian TR. Environmental transmission of diarrheal pathogens in low and middle income countries. ENVIRONMENTAL SCIENCE. PROCESSES & IMPACTS 2016; 18:944-955. [PMID: 27384220 DOI: 10.1039/c6em00222f] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Every year, more than half a million children die due to diarrheal diseases. Recent studies have identified the most important etiologies of diarrheal disease are enterotoxigenic and enteropathogenic E. coli, Shigella spp., rotavirus, norovirus and Cryptosporidium spp. These etiologies are unsurprisingly characterized by a combination of high shedding, high infectivity, and transmissibility through multiple environmental reservoirs. The relative importance of the transmission routes is likely site-specific. So the impact of interventions, which typically target only one or two environmental reservoirs, is likely also site-specific. The factors influencing the transmission routes most important for diarrheal disease are complex, including - at a minimum - etiology of endemic disease; and water, sanitation, and hygiene infrastructure and practices. The site-specific nature - and complexity of transmission - helps explain the observed variation in impacts of water, sanitation, and hygiene interventions. It may also render efforts to estimate or quantify global means for interventions' impacts irrelevant. The theme of this Perspective is that greater reductions in diarrheal disease transmission in LMICs can be achieved by designing interventions to interrupt the most important environmental transmission pathways. Intervention choice should be informed by site-specific conditions, most notably: diarrheal etiology and existing water, sanitation, and hygiene infrastructure and practices. The theme is discussed through the lens of the characteristics of the most important diarrheal diseases (shedding, infectivity, growth, and persistence) and the general characteristics of environmental reservoirs (exposure pathways and fecal contamination). The discussion highlights when interventions - and combinations of interventions - will be most effective at reducing diarrheal disease burden.
Collapse
Affiliation(s)
- Timothy R Julian
- Pathogens and Human Health, Department of Environmental Microbiology, Swiss Federal Research Institute of Aquatic Science and Technology, Eawag, BU-F08, Überlandstrasse 133, 8600 Dübendorf, Switzerland.
| |
Collapse
|
49
|
Pop M, Paulson JN, Chakraborty S, Astrovskaya I, Lindsay BR, Li S, Bravo HC, Harro C, Parkhill J, Walker AW, Walker RI, Sack DA, Stine OC. Individual-specific changes in the human gut microbiota after challenge with enterotoxigenic Escherichia coli and subsequent ciprofloxacin treatment. BMC Genomics 2016; 17:440. [PMID: 27277524 PMCID: PMC4898365 DOI: 10.1186/s12864-016-2777-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 05/25/2016] [Indexed: 01/01/2023] Open
Abstract
Background Enterotoxigenic Escherichia coli (ETEC) is a major cause of diarrhea in inhabitants from low-income countries and in visitors to these countries. The impact of the human intestinal microbiota on the initiation and progression of ETEC diarrhea is not yet well understood. Results We used 16S rRNA (ribosomal RNA) gene sequencing to study changes in the fecal microbiota of 12 volunteers during a human challenge study with ETEC (H10407) and subsequent treatment with ciprofloxacin. Five subjects developed severe diarrhea and seven experienced few or no symptoms. Diarrheal symptoms were associated with high concentrations of fecal E. coli as measured by quantitative culture, quantitative PCR, and normalized number of 16S rRNA gene sequences. Large changes in other members of the microbiota varied greatly from individual to individual, whether or not diarrhea occurred. Nonetheless the variation within an individual was small compared to variation between individuals. Ciprofloxacin treatment reorganized microbiota populations; however, the original structure was largely restored at one and three month follow-up visits. Conclusion Symptomatic ETEC infections, but not asymptomatic infections, were associated with high fecal concentrations of E. coli. Both infection and ciprofloxacin treatment caused variable changes in other bacteria that generally reverted to baseline levels after three months. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2777-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mihai Pop
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, USA.,Department of Computer Science, University of Maryland, College Park, MD, USA
| | - Joseph N Paulson
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, USA.,Graduate Program in Applied Mathematics & Scientific Computation, University of Maryland, College Park, MD, USA.,Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, Boston, MA, USA.,Department of Biostatistics, Harvard School of Public Health, Boston, MA, USA
| | | | - Irina Astrovskaya
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, USA
| | - Brianna R Lindsay
- School of Medicine, University of Maryland, Baltimore, MD, USA.,Merck & Co. Inc, North Wales, PA, USA
| | - Shan Li
- School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Héctor Corrada Bravo
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, USA.,Department of Computer Science, University of Maryland, College Park, MD, USA
| | - Clayton Harro
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Julian Parkhill
- Pathogen Genomics Group, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Alan W Walker
- Pathogen Genomics Group, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK.,Microbiology Group, Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, UK
| | | | - David A Sack
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - O Colin Stine
- School of Medicine, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
50
|
Tennant SM, Steele AD, Pasetti MF. Highlights of the 8th International Conference on Vaccines for Enteric Diseases: the Scottish Encounter To Defeat Diarrheal Diseases. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:272-81. [PMID: 26936100 PMCID: PMC4820512 DOI: 10.1128/cvi.00082-16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Infectious diarrhea is a leading cause of morbidity and of mortality; the burden of disease affects individuals of all ages but particularly young children, especially those living in poor regions where the disease is endemic. It is also a health concern for international travelers to these areas. Experts on vaccines and enteric infections and advocates for global health improvement gathered in Scotland from 8 to 10 July 2015 to discuss recent advances in the assessment and understanding of the burden of enteric diseases and progress in the development and implementation of strategies to prevent these infections. Highlights of the meeting included description of advances in molecular assays to estimate pathogen-specific prevalence, methods to model epidemiologic trends, novel approaches to generate broad-spectrum vaccines, new initiatives to evaluate vaccine performance where they are most needed, renewed interest in human challenge models, immunological readouts as predictors of vaccine efficacy, maternal immunization to prevent enteric infections, and the impact of maternal immunity on the vaccine take of infants. A follow-up scientific gathering to advance Shigella and enterotoxigenic Escherichia coli (ETEC) vaccine efforts will be held from 28 to 30 June 2016 in Washington, DC.
Collapse
Affiliation(s)
- Sharon M Tennant
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - A Duncan Steele
- Enteric and Diarrheal Diseases, Bill & Melinda Gates Foundation, Seattle, Washington, USA
| | - Marcela F Pasetti
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|