1
|
Carreto-Binaghi LE, Sztein MB, Booth JS. Role of cellular effectors in the induction and maintenance of IgA responses leading to protective immunity against enteric bacterial pathogens. Front Immunol 2024; 15:1446072. [PMID: 39324143 PMCID: PMC11422102 DOI: 10.3389/fimmu.2024.1446072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
The mucosal immune system is a critical first line of defense to infectious diseases, as many pathogens enter the body through mucosal surfaces, disrupting the balanced interactions between mucosal cells, secretory molecules, and microbiota in this challenging microenvironment. The mucosal immune system comprises of a complex and integrated network that includes the gut-associated lymphoid tissues (GALT). One of its primary responses to microbes is the secretion of IgA, whose role in the mucosa is vital for preventing pathogen colonization, invasion and spread. The mechanisms involved in these key responses include neutralization of pathogens, immune exclusion, immune modulation, and cross-protection. The generation and maintenance of high affinity IgA responses require a delicate balance of multiple components, including B and T cell interactions, innate cells, the cytokine milieu (e.g., IL-21, IL-10, TGF-β), and other factors essential for intestinal homeostasis, including the gut microbiota. In this review, we will discuss the main cellular components (e.g., T cells, innate lymphoid cells, dendritic cells) in the gut microenvironment as mediators of important effector responses and as critical players in supporting B cells in eliciting and maintaining IgA production, particularly in the context of enteric infections and vaccination in humans. Understanding the mechanisms of humoral and cellular components in protection could guide and accelerate the development of more effective mucosal vaccines and therapeutic interventions to efficiently combat mucosal infections.
Collapse
Affiliation(s)
- Laura E Carreto-Binaghi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Laboratorio de Inmunobiologia de la Tuberculosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Tumor Immunology and Immunotherapy Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Jayaum S Booth
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
2
|
Talaat KR, Porter CK, Chakraborty S, Feijoo BL, Brubaker J, Adjoodani BM, DeNearing B, Prouty MG, Poole ST, Bourgeois AL, Billingsley M, Sack DA, Eder-Lingelbach S, Taucher C. Validation of a Human Challenge Model Using an LT-Expressing Enterotoxigenic E. coli Strain (LSN03-016011) and Characterization of Potential Amelioration of Disease by an Investigational Oral Vaccine Candidate (VLA1701). Microorganisms 2024; 12:727. [PMID: 38674674 PMCID: PMC11051778 DOI: 10.3390/microorganisms12040727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/22/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024] Open
Abstract
Controlled human infection models are important tools for the evaluation of vaccines against diseases where an appropriate correlate of protection has not been identified. Enterotoxigenic Escherichia coli (ETEC) strain LSN03-016011/A (LSN03) is an LT enterotoxin and CS17-expressing ETEC strain useful for evaluating vaccine candidates targeting LT-expressing strains. We sought to confirm the ability of the LSN03 strain to induce moderate-to-severe diarrhea in a healthy American adult population, as well as the impact of immunization with an investigational cholera/ETEC vaccine (VLA-1701) on disease outcomes. A randomized, double-blinded pilot study was conducted in which participants received two doses of VLA1701 or placebo orally, one week apart; eight days after the second vaccination, 30 participants (15 vaccinees and 15 placebo recipients) were challenged with approximately 5 × 109 colony-forming units of LSN03. The vaccine was well tolerated, with no significant adverse events. The vaccine also induced serum IgA and IgG responses to LT. After challenge, 11 of the placebo recipients (73.3%; 95%CI: 48.0-89.1) and 7 of the VLA1701 recipients (46.7%; 95%CI: 24.8-68.8) had moderate-to-severe diarrhea (p = 0.26), while 14 placebo recipients (93%) and 8 vaccine recipients (53.3%) experienced diarrhea of any severity, resulting in a protective efficacy of 42.9% (p = 0.035). In addition, the vaccine also appeared to provide protection against more severe diarrhea (p = 0.054). Vaccinees also tended to shed lower levels of the LSN03 challenge strain compared to placebo recipients (p = 0.056). In addition, the disease severity score was lower for the vaccinees than for the placebo recipients (p = 0.046). In summary, the LSN03 ETEC challenge strain induced moderate-to-severe diarrhea in 73.3% of placebo recipients. VLA1701 vaccination ameliorated disease severity, as observed by several parameters, including the percentage of participants experiencing diarrhea, as well as stool frequency and ETEC severity scores. These data highlight the potential value of LSN03 as a suitable ETEC challenge strain to evaluate LT-based vaccine targets (NCT03576183).
Collapse
Affiliation(s)
- Kawsar R. Talaat
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | - Chad K. Porter
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (C.K.P.); (M.G.P.); (S.T.P.)
| | - Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | - Brittany L. Feijoo
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | - Jessica Brubaker
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | - Brittany M. Adjoodani
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | - Barbara DeNearing
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | - Michael G. Prouty
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (C.K.P.); (M.G.P.); (S.T.P.)
| | - Steven T. Poole
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (C.K.P.); (M.G.P.); (S.T.P.)
| | - A. Louis Bourgeois
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | - Madison Billingsley
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | - David A. Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | | | | |
Collapse
|
3
|
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
|
4
|
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
|
5
|
DuPont HL, Almenoff JS, Jamindar MS, Bortey E, Steffen R. Development of a new travellers' diarrhoea clinical severity classification and its utility in confirming rifamycin-SV efficacy. J Travel Med 2023; 30:taad043. [PMID: 37052453 PMCID: PMC10628770 DOI: 10.1093/jtm/taad043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND travellers' diarrhoea (TD) is frequently reported with incidence up to 40% in high-risk destinations. Previous studies showed that the number of loose stools alone is inadequate to holistically predict the severity of TD. To improve the prediction of prognosis and to optimize treatments, a simple risk-based clinical severity classification has been developed. METHODS pooled baseline data of signs and symptoms and number of loose stools from 1098 subjects enrolled in two double-blind Phase 3 trials of rifamycin-SV were analyzed with correlation, multiple correspondence analyses, prognostic factor criteria, and Contal and O'Quigley method to generate a TD severity classification (mild, moderate and severe). The relative importance of this classification on resolution of TD was assessed by Cox proportional model hazard model on the time to last unformed stool (TLUS). RESULTS the analysis showed that TLUS were longer for the severe [hazard ratio (HR) 0.24; P < 0.001; n = 173] and moderate (HR 0.54; P = 0.0272; n = 912) vs mild. Additionally, when the treatment assigned in the studies was investigated in the severity classification, the results yielded that rifamycin-SV significantly shortened TLUS vs placebo for all subjects (HR 1.9; P = 0.0006), severe (HR 5.9; P = 0.0232) and moderate (HR 1.7; P = 0.0078) groups and was as equally efficacious as ciprofloxacin for all subjects, moderate and severe groups (HRs: 0.962, 0.9, 1.2; all P = NS, respectively). When reassessed by this classification, rifamycin-SV showed consistent efficacy with the Phase 3 studies. CONCLUSIONS this newly developed TD clinical severity classification demonstrated strong prognostic value and clinical utility by combining patients' multiple signs and symptoms of enteric infection and number of loose stools to provide a holistic assessment of TD. By expanding on the current classification by incorporating patient reported outcomes in addition to TLUS, a classification like the one developed, may help optimize patient selection for future clinical studies.
Collapse
Affiliation(s)
- Herbert L DuPont
- Internal Medicine, University of Texas School of Public Health, Houston, TX 77030, USA
| | - June S Almenoff
- Department of Medical Affairs, Redhill Biopharma Inc., Raleigh, NC 27617, USA
| | - Mansi S Jamindar
- Department of Medical Affairs, Redhill Biopharma Inc., Raleigh, NC 27617, USA
| | - Enoch Bortey
- Pharmaceutical Development Strategies LLC, Chapel Hill, NC 27517, USA
| | - Robert Steffen
- Department of Epidemiology, University of Zurich, 8001 Zurich, Switzerland
| |
Collapse
|
6
|
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: 4.0] [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
|
7
|
Mantua J, Gutierrez RL, Isidean SD, Alcala AN, Testa KJ, Talaat KR, Doty TJ, Porter CK. Sleep duration prior to an enterotoxigenic Escherichia coli challenge predicts diarrhea severity during infection. Sleep Med 2022; 100:404-409. [PMID: 36240601 DOI: 10.1016/j.sleep.2022.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 09/09/2022] [Accepted: 09/11/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Acute diarrhea is the most frequent diagnosis among ill travelers. Sleep loss may weaken the body's defense against pathogens and increase susceptibility to infection. The relationship between sleep and infectious diarrhea has not been studied and was assessed utilizing data from a controlled human infection model (CHIM) for enterotoxigenic Escherichia coli (ETEC). METHODS During a CHIM assessing the efficacy of an immunoprophylactic targeting ETEC against moderate-to-severe diarrhea (MSD) following challenge, we measured sleep via actigraphy over an 8-day inpatient period. We hypothesized better sleep pre-challenge would predict illness symptomatology following challenge. RESULTS Among 57 participants (aged 34.4 ± 8.1 years, 64% male), there was no relationship between sleep metrics and incidence of MSD. However, longer total sleep time the night preceding ETEC challenge was associated with lower maximum 24 h diarrhea volume (B = -1.80, p = 0.01) and total diarrhea volume (B = -2.45, p = 0.01). CONCLUSIONS This novel study showed that shorter sleep duration predicted diarrhea severity over the course of an ETEC infection. Future work should experimentally manipulate sleep to further clarify its impact on diarrhea-related outcomes for ETEC and other important enteric pathogens.
Collapse
Affiliation(s)
- Janna Mantua
- Walter Reed Army Institute of Research, Behavioral Biology Branch, Silver Spring, MD, 20910, USA.
| | - Ramiro L Gutierrez
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, 20910, USA
| | - Sandra D Isidean
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Ashley N Alcala
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Kayla J Testa
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Kawsar R Talaat
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Tracy Jill Doty
- Walter Reed Army Institute of Research, Behavioral Biology Branch, Silver Spring, MD, 20910, USA
| | - Chad K Porter
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, 20910, USA
| |
Collapse
|
8
|
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: 11] [Impact Index Per Article: 5.5] [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
|
9
|
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: 12] [Impact Index Per Article: 6.0] [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
|
10
|
Maier N, Riddle MS, Gutiérrez R, Fraser JA, Connor P, Tribble DR, Porter CK. A disease severity scale for the evaluation of vaccine and other preventive or therapeutic interventions for travellers' diarrhoea. J Travel Med 2022; 29:6365136. [PMID: 34490456 PMCID: PMC8763125 DOI: 10.1093/jtm/taab139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 11/14/2022]
Abstract
BACKGROUND Travellers' diarrhoea (TD) is the most common travel-related illness with an estimated 10 million people afflicted annually. Outcome measures to assess the efficacy of primary and secondary TD interventions were historically based on diarrhoea frequency with ≥1 associated gastrointestinal symptom. Furthermore, efficacy determination is often made on the presence or absence of TD, rather than on TD illness severity. Current severity classifications are based on subjective consideration of impact of illness on activity. We sought to develop a standardized scoring system to characterize TD severity to potentially apply as a secondary outcome in future field studies. METHODS Data on multiple signs and symptoms were obtained from a previously published multisite TD treatment trial conducted by the US Department of Defense (TrEAT TD). Correlation, regression and multiple correspondence analyses were performed to assess impact on activity and a TD severity score was established. RESULTS Numerous signs and symptoms were associated with impaired function, with malaise and nausea most strongly associated [odds ratio (OR) 5.9-44.3, P < 0.0001 and OR 2.8-37.1, P < 0.0001, respectively). Based on co-varying symptomatology, a TD severity score accounting for diarrhoea frequency in addition to several signs and symptoms was a better predictor of negative impact on function than any single sign/symptom (X2 = 127.16, P < 0.001). Additionally, there was a significant difference (P < 0.0001) in the mean TD severity score between those with acute watery diarrhoea (3.9 ± 1.9) and those with dysentery or acute febrile illness (6.2 ± 2.0). CONCLUSIONS The newly developed disease severity score better predicted a negative impact on activity due to TD than did any single sign or symptom. Incorporating multiple parameters into the TD severity score better captures illness severity and moves the field towards current recommendations for TD management by considering symptoms with high functional impact. Further validation of this score is needed in non-military travellers and other settings.
Collapse
|
11
|
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: 3] [Impact Index Per Article: 0.8] [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
|
12
|
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: 1.3] [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
|
13
|
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: 0.8] [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
|
14
|
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: 3.5] [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
|
15
|
Hosangadi D, Smith PG, Kaslow DC, Giersing BK. WHO consultation on ETEC and Shigella burden of disease, Geneva, 6–7th April 2017: Meeting report. Vaccine 2019; 37:7381-7390. [DOI: 10.1016/j.vaccine.2017.10.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 10/03/2017] [Indexed: 10/18/2022]
|
16
|
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: 3.2] [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
|
17
|
Ena J, Afonso-Carrillo RG, Bou-Collado M, Galian-Nicolas V, Reyes-Jara MD, Martínez-Peinado C, Gomez-Alonso B, Arjona-Zaragozi F. Epidemiology of Severe Acute Diarrhea in Patients Requiring Hospital Admission. J Emerg Med 2019; 57:290-298. [DOI: 10.1016/j.jemermed.2019.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/06/2019] [Accepted: 06/08/2019] [Indexed: 01/21/2023]
|
18
|
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: 2.2] [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
|
19
|
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: 1.6] [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
|
20
|
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: 2.0] [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
|
21
|
Establishment, Validation, and Application of a New World Primate Model of Enterotoxigenic Escherichia coli Disease for Vaccine Development. Infect Immun 2019; 87:IAI.00634-18. [PMID: 30510102 DOI: 10.1128/iai.00634-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/19/2018] [Indexed: 12/27/2022] Open
Abstract
The establishment of an animal model that closely approximates enterotoxigenic Escherichia coli (ETEC) disease in humans is critical for the development and evaluation of vaccines against this enteropathogen. Here, we evaluated the susceptibility of Aotus nancymaae, a New World monkey species, to ETEC infection. Animals were challenged orogastrically with 109 to 1011 CFU of the human pathogenic CFA/I+ ETEC strain H10407 and examined for evidence of diarrhea and fecal shedding of bacteria. A clear dose-range effect was obtained, with diarrheal attack rates of 40% to 80%, validated in a follow-on study demonstrating an attack rate of 80% with 1011 CFU of H10407 ETEC. To determine whether this model is an effective approach for assessing ETEC vaccine candidates, we used it to evaluate the ability of the donor strand-complemented CFA/I adhesin CfaE (dscCfaE) to protect against H10407 challenge. In a series of experiments, animals were intranasally vaccinated with dscCfaE alone, dscCfaE with either cholera toxin B-subunit (CTB) or heat-labile toxin (LTB), or phosphate-buffered saline (PBS) alone and then challenged with 1011 CFU of H10407. Control animals vaccinated with PBS had attack rates of 70 to 90% on challenge. Vaccination with dscCfaE, or dscCfaE admixed with CTB or LTB, resulted in a reduction of attack rates, with vaccine efficacies of 66.7% (P = 0.02), 77.7% (P = 0.006), and 42.9% (P = 0.370) to 83.3% (P = 0.041), respectively. In conclusion, we have shown the H10407 ETEC challenge of A. nancymaae to be an effective, reproducible model of ETEC disease, and importantly, we have demonstrated that in this model, vaccination with the prototype vaccine candidate dscCfaE is protective against CF-homologous disease.
Collapse
|
22
|
Caron P, Brue T, Raverot G, Tabarin A, Cailleux A, Delemer B, Renoult PP, Houchard A, Elaraki F, Chanson P. Signs and symptoms of acromegaly at diagnosis: the physician's and the patient's perspectives in the ACRO-POLIS study. Endocrine 2019; 63:120-129. [PMID: 30269264 PMCID: PMC6329724 DOI: 10.1007/s12020-018-1764-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 09/15/2018] [Indexed: 01/16/2023]
Abstract
PURPOSE Acromegaly is characterized by a broad range of manifestations. Early diagnosis is key to treatment success, but is often delayed as symptomatology overlaps with common disorders. We investigated sign-and-symptom associations, demographics, and clinical characteristics at acromegaly diagnosis. METHODS Observational, cross-sectional, multicenter non-interventional study conducted at 25 hospital departments in France that treat acromegaly (ClinicalTrials.gov: NCT02012127). Adults diagnosed with acromegaly < 5 years were enrolled. Demographic and clinical data were obtained from medical reports and patient questionnaires. Sign-and-symptom associations were assessed by multiple correspondence analysis (MCA). RESULTS Overall, 472 patients were included in the analyses. MCA was unsuccessful in identifying sign-and-symptom associations at diagnosis. Endocrinologists (29.5% patients) and other clinical specialists (37.2% patients) were commonly first to suspect acromegaly. Morphologic manifestations (83.7-87.9% patients), snoring syndrome (81.4% patients), and asthenia (79.2% patients) were frequently present at diagnosis; differences were found between sexes for specific manifestations. Rates of discrepancy between patient- and physician-reported manifestations were highest for functional signs. Earliest manifestations prior to diagnosis, according to how they were detected, were enlarged hands and feet (6.4 ± 6.8 and 6.2 ± 6.9 years, functional signs), hypertension (6.6 ± 7.5 years, complementary examination) and carpal/cubital tunnel syndrome (5.7 ± 6.7 years, functional signs with complementary examination). CONCLUSIONS Results confirm the broad range of manifestations at diagnosis and delay in recognizing the disease. We identified early manifestations and sex differences that may aid physicians in diagnosing acromegaly. Discrepancy rates suggest physicians should obtain the patient's perspective and seek functional signs during diagnosis.
Collapse
Affiliation(s)
| | - Thierry Brue
- Aix-Marseille University, INSERM, MMG, AP-HM, Hôpital de la Conception, CRMR HYPO, Marseille, France
| | | | | | - Anne Cailleux
- Rouen University Hospital, Endocrinology Unit, Inserm CIC-CRB 1404, F 76 000, Rouen, France
| | | | | | | | | | - Philippe Chanson
- Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Centre de Référence des Maladies Rares de l'Hypophyse HYPO, F94275, Le Kremlin-Bicêtre, France.
- Université Paris-Sud, Le Kremlin-Bicêtre, France.
| |
Collapse
|
23
|
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: 6.3] [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
|
24
|
Porter CK, Lynen A, Riddle MS, Talaat K, Sack D, Gutiérrez RL, McKenzie R, DeNearing B, Feijoo B, Kaminski RW, Taylor DN, Kirkpatrick BD, Bourgeois AL. Clinical endpoints in the controlled human challenge model for Shigella: A call for standardization and the development of a disease severity score. PLoS One 2018; 13:e0194325. [PMID: 29590182 PMCID: PMC5874036 DOI: 10.1371/journal.pone.0194325] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 02/28/2018] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Since 1946 the controlled human infection model (CHIM) for Shigella has been used to improve understanding of disease pathogenesis, describe clinical and immunologic responses to infection and as a tool for vaccine development. As the frequency and intent for use in vaccine comparisons increases, standardization of the primary endpoint definition is necessary. METHODS Subject-level data were obtained from previously conducted experimental Shigella CHIM studies. Signs and symptoms severity were categorized consistently across all studies. Sign and symptom correlations were estimated and univariate models were utilized to describe the association between stool output and other Shigella-attributable signs and symptoms. Multiple correspondence and hierarchical clustering analyses were performed to describe the co-occurrence of signs and symptoms. A disease score is proposed based on the co-occurrence of these events. RESULTS Data were obtained on 54 subjects receiving 800 to 2000 colony forming units (cfu) of S. flexneri. The median maximum 24 hour stool output was 514 ml (IQR: 300, 998 ml) with a median frequency of 6 (IQR: 4, 9). Subjects reported abdominal pain or cramps (81.5%), headache (66.7%) and anorexia (64.8%), 50.0% had a fever and 27.8% had gross blood in multiple loose stools. Multiple correspondence analyses highlighted co-occurrence of symptoms based on severity. A 3-parameter disease severity score predicted shigellosis endpoints and better differentiated disease spectrum. CONCLUSION Dichotomous endpoints for Shigella CHIM fail to fully account for disease variability. An ordinal disease score characterizing the breadth of disease severity may enable a better characterization of shigellosis and can decrease sample size requirements. Furthermore, the disease severity score may be a useful tool for portfolio management by enabling prioritization across vaccine candidates with comparable efficacy estimates using dichotomous endpoints.
Collapse
Affiliation(s)
- Chad K. Porter
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, MD, United States of America
- * E-mail:
| | - Amanda Lynen
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Mark S. Riddle
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Kawsar Talaat
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - David Sack
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Ramiro L. Gutiérrez
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Robin McKenzie
- School of Medicine, Johns Hopkins University, Baltimore, MD, United States of America
| | - Barbara DeNearing
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Brittany Feijoo
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Robert W. Kaminski
- Department of Enteric Infections, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - David N. Taylor
- Drug Development Global Program, PATH, Seattle, WA, United States of America
| | - Beth D. Kirkpatrick
- University of Vermont College of Medicine, Vaccine Testing Center, Department of Medicine, Burlington, VT, United States of America
| | - A. Louis Bourgeois
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
- Enteric Vaccine Initiative, PATH, Washington, DC, United States of America
| |
Collapse
|
25
|
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: 4.3] [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
|
26
|
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: 56] [Impact Index Per Article: 9.3] [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
|
27
|
Khan F, Srivastava V, Kumar A. Computational Identification and Characterization of Potential T-Cell Epitope for the Utility of Vaccine Design Against Enterotoxigenic Escherichia coli. Int J Pept Res Ther 2018. [DOI: 10.1007/s10989-018-9671-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
28
|
Porter CK, Louis Bourgeois A, Frenck RW, Prouty M, Maier N, Riddle MS. Developing and utilizing controlled human models of infection. Vaccine 2017; 35:6813-6818. [PMID: 28583306 DOI: 10.1016/j.vaccine.2017.05.068] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 04/11/2017] [Accepted: 05/24/2017] [Indexed: 11/18/2022]
Abstract
The controlled human infection model (CHIM) to assess the efficacy of vaccines against Shigella and enterotoxigenic Escherichia coli (ETEC) has several unique features that could significantly enhance the ability to test candidate vaccines. Despite increasing interest in these models, questions remain as to how to best incorporate them into vaccine development and how to maximize results. We designed a workshop focused on CHIM as part of the Vaccines Against Shigella and ETEC (VASE) Conference. The workshop, using the World Café method, focused on; clinical outcomes, nonclinical outcomes and model standardization. Researchers with a variety of expertise and experience rotated through each focus area and discussed relevant sub-topics. The results of these discussions were presented and questions posed to guide future workshops. Clinical endpoint discussions focused on the need for harmonized definitions; optimized attack rates; difficulties of sample collection and a need for non-stool based endpoints. Nonclinical discussions centered on evolving omics-based opportunities, host predictors of susceptibility and novel characterizations of the immune response. Model standardization focused on the value of shared procedures across institutions for clinical and non-clinical endpoints as well as for strain preparation and administration and subject selection. Participants agreed CHIMs for Shigella and ETEC vaccine development could accelerate vaccine development of a promising candidate; however, it was also appreciated that variability in the model and our limited understand of the host-pathogen interaction may yield results that could negatively impact a suitable candidate. Future workshops on CHIM are needed to ensure the optimal application of these models moving forward.
Collapse
Affiliation(s)
- Chad K Porter
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, United States.
| | - A Louis Bourgeois
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Robert W Frenck
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Michael Prouty
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, United States
| | | | - Mark S Riddle
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, United States
| |
Collapse
|
29
|
Delucchi L, Fraga M, Zunino P. Effect of the probiotic Lactobacillus murinus LbP2 on clinical parameters of dogs with distemper-associated diarrhea. CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2017; 81:118-121. [PMID: 28408779 PMCID: PMC5370537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 09/22/2016] [Indexed: 06/07/2023]
Abstract
The objective of this study was to assess the effect of the probiotic Lactobacillus murinus native strain (LbP2) on general clinical parameters of dogs with distemper-associated diarrhea. Two groups of dogs over 60 d of age with distemper and diarrhea were used in the study, which was done at the Animal Hospital of the Veterinary Faculty of the University of Uruguay, Montevideo, Uruguay. The dogs were treated orally each day for 5 d with the probiotic or with a placebo (vehicle without bacteria). Clinical parameters were assessed and scored according to a system specially designed for this study. Blood parameters were also measured. Administration of the probiotic significantly improved the clinical score of the patients, whereas administration of the placebo did not. Stool output, fecal consistency, mental status, and appetite all improved in the probiotic-treated dogs. These results support previous findings of beneficial effects with the probiotic L. murinus LbP2 in dogs. Thus, combined with other therapeutic measures, probiotic treatment appears to be promising for the management of canine distemper-associated diarrhea.
Collapse
Affiliation(s)
| | | | - Pablo Zunino
- Address all correspondence to Dr. Pablo Zunino; telephone: 598-24871616; fax: 598-24875461; e-mail:
| |
Collapse
|
30
|
McArthur MA, Chen WH, Magder L, Levine MM, Sztein MB. Impact of CD4+ T Cell Responses on Clinical Outcome following Oral Administration of Wild-Type Enterotoxigenic Escherichia coli in Humans. PLoS Negl Trop Dis 2017; 11:e0005291. [PMID: 28103236 PMCID: PMC5283752 DOI: 10.1371/journal.pntd.0005291] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/31/2017] [Accepted: 12/29/2016] [Indexed: 12/24/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a non-invasive enteric pathogen of considerable public health importance, being one of the most common attributable causes of diarrheal illness in infants and young children in developing countries and the most common cause of traveler’s diarrhea. To enhance study-to-study consistency of our experimental challenge model of ETEC in volunteers, and to allow concomitant multi-site trials to evaluate anti-ETEC immunoprophylactic products, hundreds of vials, each containing a standardized inoculum of virulent wild-type (wt) ETEC strain H10407 (serotype O78:H11 expressing colonization factor antigen I and heat-labile and heat-stable enterotoxins), were prepared under current Good Manufacturing Practices (cGMP) and frozen. Following thawing, the contents of each vial can be used (diluted as necessary) to prepare consistent challenge inoculum, even at different study sites. A preliminary human experimental challenge study using this cGMP inoculum was conducted on a research isolation ward and the clinical and cell-mediated immune responses evaluated. Of the 6 healthy adult volunteers challenged 83% (5/6) developed diarrhea and 50% developed moderate-to-severe diarrhea (MSD). Moderate and severe diarrhea were defined as passage of ≥ 1 liter or ≥ 3 liters of diarrheal stool respectively. We compared the CD4+ T cell responses of volunteers who developed MSD against those who did not and identified significant differences in ETEC-specific cytokine production and gut homing potential. We furthermore demonstrated that increased expression of the gut-homing molecule integrin α4β7 by peripheral T follicular helper cells (pTfh) correlated with decreased stool volume and increased ETEC-specific IgA B memory cell (BM) development. Collectively, despite small numbers of volunteers, our results indicate a potential role for CD4+ T cells, in particular pTfh, in modulating disease outcome following exposure to wt ETEC in a volunteer experimental challenge model. Enterotoxigenic Escherichia coli (ETEC) is an important cause of diarrheal illness in infants and young children in the developing world, as well as in individuals traveling to endemic areas. Due to the lack of suitable animal models for human ETEC infection, we performed a human challenge study in which volunteers ingested wild-type ETEC in a controlled clinical setting. In addition to closely monitoring their clinical status, we studied their ETEC-specific T cell responses prior to and after challenge and studied the presence of associations between CD4+ T cell responses and clinical outcome. We observed differences in the immunological responses of individuals who developed moderate to severe diarrhea following challenge compared to those who did not. These results indicate that T cells may be an important component of the immune response against ETEC.
Collapse
Affiliation(s)
- Monica A. McArthur
- Center for Vaccine Development (CVD), University of Maryland, Baltimore, Maryland, United States of America
- * E-mail:
| | - Wilbur H. Chen
- Center for Vaccine Development (CVD), University of Maryland, Baltimore, Maryland, United States of America
| | - Laurence Magder
- Department of Epidemiology and Public Health, University of Maryland, Baltimore, Maryland, United States of America
| | - Myron M. Levine
- Center for Vaccine Development (CVD), University of Maryland, Baltimore, Maryland, United States of America
| | - Marcelo B. Sztein
- Center for Vaccine Development (CVD), University of Maryland, Baltimore, Maryland, United States of America
| |
Collapse
|