101
|
Yang Z, Shi L, Yu H, Zhang Y, Chen K, Saint Fleur A, Bai G, Feng H. Intravenous adenovirus expressing a multi-specific, single-domain antibody neutralizing TcdA and TcdB protects mice from Clostridium difficile infection. Pathog Dis 2016; 74:ftw078. [PMID: 27502696 DOI: 10.1093/femspd/ftw078] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2016] [Indexed: 01/09/2023] Open
Abstract
Clostridium difficile infection (CDI) is the most common cause of antibiotic-associated diarrhea and colitis in developed countries. The disease is mainly mediated via two major exotoxins TcdA and TcdB secreted by the bacterium. We have previously developed a novel, potently neutralizing, tetravalent and bispecific heavy-chain-only single domain (VHH) antibody to both TcdA and TcdB (designated as ABA) that reverses fulminant CDI in mice. Since ABA has a short serum half-life, in this study a replication-deficient recombinant adenovirus expressing ABA was generated and the long-lasting expression of functional ABA was demonstrated in vitro and in vivo Mice transduced with one dose of the adenovirus displayed high levels of serum ABA for more than1 month and were fully protected against systemic toxin challenges. More importantly, the ABA delivered by the adenovirus protected mice from both primary and recurrent CDI. Thus, replication-deficient adenoviral vector may be used to deliver neutralizing antibodies against the toxins in order to prevent CDI and recurrence.
Collapse
Affiliation(s)
- Zhiyong Yang
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Lianfa Shi
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Hua Yu
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Yongrong Zhang
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Kevin Chen
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Ashley Saint Fleur
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Guang Bai
- Department of Neural and Pain Sciences, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Hanping Feng
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| |
Collapse
|
102
|
Abstract
Clostridium difficile continues to be one of the most prevalent hospital-acquired bacterial infections in the developed world, despite the recent introduction of a novel and effective antibiotic agent (fidaxomicin). Alternative approaches under investigation to combat the anaerobic Gram-positive bacteria include fecal transplantation therapy, vaccines, and antibody-based immunotherapies. In this review, we catalog the recent advances in antibody-based approaches under development and in the clinic for the treatment of C. difficile infection. By and large, inhibitory antibodies that recognize the primary C. difficile virulence factors, toxin A and toxin B, are the most popular passive immunotherapies under investigation. We provide a detailed summary of the toxin epitopes recognized by various antitoxin antibodies and discuss general trends on toxin inhibition efficacy. In addition, antibodies to other C. difficile targets, such as surface-layer proteins, binary toxin, motility factors, and adherence and colonization factors, are introduced in this review.
Collapse
Affiliation(s)
- Greg Hussack
- Human Health Therapeutics Portfolio, National Research Council Canada, Ottawa
| | - Jamshid Tanha
- Human Health Therapeutics Portfolio, National Research Council Canada, Ottawa; School of Environmental Sciences, University of Guelph, Guelph; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
103
|
Gupta SB, Mehta V, Dubberke ER, Zhao X, Dorr MB, Guris D, Molrine D, Leney M, Miller M, Dupin M, Mast TC. Antibodies to Toxin B Are Protective Against Clostridium difficile Infection Recurrence. Clin Infect Dis 2016; 63:730-734. [PMID: 27365387 DOI: 10.1093/cid/ciw364] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 05/07/2016] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Although newer studies have evaluated risk factors for recurrent Clostridium difficile infection (CDI), the vast majority did not measure important biomarkers such as endogenous anti-toxin A and anti-toxin B antibody levels. METHODS Data from the placebo group of a phase 2 trial testing monoclonal antibodies to C. difficile toxins A and B for preventing CDI recurrence (rCDI) were analyzed to assess risk factors associated with rCDI. Patients with symptomatic CDI taking metronidazole or vancomycin were enrolled. The primary outcome was rCDI within 84 days of treatment start. Univariate and multivariate logistic regression was used to examine associations between potential risk factors and rCDI. At baseline, demographic and clinical characteristics were recorded; endogenous antibody levels were assessed using 2 enzyme-linked immunosorbent assays. RESULTS A predictor of recurrence was age ≥65 years, and an antibody-mediated immune response to toxin B appears to be protective against rCDI. CONCLUSIONS Our findings demonstrate the importance of clinical as well as immunological risk factors in rCDI and provide more robust evidence for the protective effects of antibody to toxin B in the prevention of rCDI. CLINICAL TRIALS REGISTRATION NCT00350298.
Collapse
Affiliation(s)
| | - Vinay Mehta
- Pharmacoepidemiology, Merck & Co, Inc, Kenilworth, New Jersey
| | - Erik R Dubberke
- Division of Infectious Diseases, Washington University School of Medicine, St Louis, Missouri
| | | | - Mary Beth Dorr
- Clinical Research, Merck & Co, Inc, Kenilworth, New Jersey
| | - Dalya Guris
- Clinical Research, Merck & Co, Inc, Kenilworth, New Jersey
| | - Deborah Molrine
- Department of Pediatrics, University of Massachusetts Medical School, Worcester.,MassBiologics, Boston
| | - Mark Leney
- MassBiologics, Boston.,Department of Medicine, University of Massachusetts Medical School, Worcester
| | | | - Marilyne Dupin
- Medical Diagnostics Discovery Department, bioMérieux, Marcy L'Etoile, France
| | | |
Collapse
|
104
|
Cornely OA, Vehreschild MJGT. Editorial Commentary: Where to Place the New Treatments for Clostridium difficile Infection? Clin Infect Dis 2016; 63:735-6. [PMID: 27365386 DOI: 10.1093/cid/ciw366] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 05/26/2016] [Indexed: 11/13/2022] Open
Affiliation(s)
- Oliver A Cornely
- Department I of Internal Medicine and Center for Integrated Oncology CIO Köln/Bonn, University Hospital of Cologne German Centre for Infection Research, Partner Site Bonn-Cologne Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, and Clinical Trials Center Cologne, ZKS Köln, University of Cologne, Germany
| | - Maria J G T Vehreschild
- Department I of Internal Medicine and Center for Integrated Oncology CIO Köln/Bonn, University Hospital of Cologne German Centre for Infection Research, Partner Site Bonn-Cologne
| |
Collapse
|
105
|
Qiu H, Cassan R, Johnstone D, Han X, Joyee AG, McQuoid M, Masi A, Merluza J, Hrehorak B, Reid R, Kennedy K, Tighe B, Rak C, Leonhardt M, Dupas B, Saward L, Berry JD, Nykiforuk CL. Novel Clostridium difficile Anti-Toxin (TcdA and TcdB) Humanized Monoclonal Antibodies Demonstrate In Vitro Neutralization across a Broad Spectrum of Clinical Strains and In Vivo Potency in a Hamster Spore Challenge Model. PLoS One 2016; 11:e0157970. [PMID: 27336843 PMCID: PMC4919053 DOI: 10.1371/journal.pone.0157970] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 06/08/2016] [Indexed: 01/07/2023] Open
Abstract
Clostridium difficile (C. difficile) infection (CDI) is the main cause of nosocomial antibiotic-associated colitis and increased incidence of community-associated diarrhea in industrialized countries. At present, the primary treatment of CDI is antibiotic administration, which is effective but often associated with recurrence, especially in the elderly. Pathogenic strains produce enterotoxin, toxin A (TcdA), and cytotoxin, toxin B (TcdB), which are necessary for C. difficile induced diarrhea and gut pathological changes. Administration of anti-toxin antibodies provides an alternative approach to treat CDI, and has shown promising results in preclinical and clinical studies. In the current study, several humanized anti-TcdA and anti-TcdB monoclonal antibodies were generated and their protective potency was characterized in a hamster infection model. The humanized anti-TcdA (CANmAbA4) and anti-TcdB (CANmAbB4 and CANmAbB1) antibodies showed broad spectrum in vitro neutralization of toxins from clinical strains and neutralization in a mouse toxin challenge model. Moreover, co-administration of humanized antibodies (CANmAbA4 and CANmAbB4 cocktail) provided a high level of protection in a dose dependent manner (85% versus 57% survival at day 22 for 50 mg/kg and 20 mg/kg doses, respectively) in a hamster gastrointestinal infection (GI) model. This study describes the protective effects conferred by novel neutralizing anti-toxin monoclonal antibodies against C. difficile toxins and their potential as therapeutic agents in treating CDI.
Collapse
Affiliation(s)
- Hongyu Qiu
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Robyn Cassan
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Darrell Johnstone
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Xiaobing Han
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Antony George Joyee
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Monica McQuoid
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Andrea Masi
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - John Merluza
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Bryce Hrehorak
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Ross Reid
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Kieron Kennedy
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Bonnie Tighe
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Carla Rak
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Melanie Leonhardt
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Brian Dupas
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Laura Saward
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Jody D. Berry
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Cory L. Nykiforuk
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| |
Collapse
|
106
|
Ross CL, Spinler JK, Savidge TC. Structural and functional changes within the gut microbiota and susceptibility to Clostridium difficile infection. Anaerobe 2016; 41:37-43. [PMID: 27180006 DOI: 10.1016/j.anaerobe.2016.05.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/05/2016] [Accepted: 05/10/2016] [Indexed: 02/06/2023]
Abstract
Alteration of the gut microbial community structure and function through antibiotic use increases susceptibility to colonization by Clostridium difficile and other enteric pathogens. However, the mechanisms that mediate colonization resistance remain elusive. As the leading definable cause of infectious diarrhea, toxigenic C. difficile represents a burden for patients and health care systems, underscoring the need for better diagnostics and treatment strategies. Next-generation sequence data has increased our understanding of how the gut microbiota is influenced by many factors including diet, disease, aging and drugs. However, a microbial-based biomarker differentiating C. difficile infection from antibiotic-associated diarrhea has not been identified. Metabolomics profiling, which is highly responsive to changes in physiological conditions, have shown promise in differentiating subtle disease phenotypes that exhibit a nearly identical microbiome community structure, suggesting metabolite-based biomarkers may be an ideal diagnostic for identifying patients with CDI. This review focuses on the current understanding of structural and functional changes to the gut microbiota during C. difficile infection obtained from studies assessing the microbiome and metabolome of samples from patients and murine models.
Collapse
Affiliation(s)
- Caná L Ross
- Texas Children's Microbiome Center, Department of Pathology, Texas Children's Hospital, 1102 Bates Ave., Houston, TX, USA; Department of Pathology & Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Jennifer K Spinler
- Texas Children's Microbiome Center, Department of Pathology, Texas Children's Hospital, 1102 Bates Ave., Houston, TX, USA; Department of Pathology & Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Tor C Savidge
- Texas Children's Microbiome Center, Department of Pathology, Texas Children's Hospital, 1102 Bates Ave., Houston, TX, USA; Department of Pathology & Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA.
| |
Collapse
|
107
|
Safety, immunogenicity and dose response of VLA84, a new vaccine candidate against Clostridium difficile, in healthy volunteers. Vaccine 2016; 34:2585-92. [DOI: 10.1016/j.vaccine.2016.03.098] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/18/2016] [Accepted: 03/30/2016] [Indexed: 12/17/2022]
|
108
|
Lees EA, Miyajima F, Pirmohamed M, Carrol ED. The role of Clostridium difficile in the paediatric and neonatal gut - a narrative review. Eur J Clin Microbiol Infect Dis 2016; 35:1047-57. [PMID: 27107991 PMCID: PMC4902830 DOI: 10.1007/s10096-016-2639-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 03/22/2016] [Indexed: 01/15/2023]
Abstract
Clostridium difficile is an important nosocomial pathogen in adults. Its significance in children is less well defined, but cases of C. difficile infection (CDI) appear to be increasingly prevalent in paediatric patients. This review aims to summarize reported Clostridium difficile carriage rates across children of different age groups, appraise the relationship between CDI and factors such as method of delivery, type of infant feed, antibiotic use, and co-morbidities, and review factors affecting the gut microbiome in children and the host immune response to C. difficile. Searches of PubMed and Google Scholar using the terms 'Clostridium difficile neonates' and 'Clostridium difficile children' were completed, and reference lists of retrieved publications screened for further papers. In total, 88 papers containing relevant data were included. There was large inter-study variation in reported C. difficile carriage rates. There was an association between CDI and recent antibiotic use, and co-morbidities such as immunosuppression and inflammatory bowel disease. C. difficile was also found in stools of children with diarrhoea attributed to other pathogens (e.g. rotavirus). The role of C. difficile in the paediatric gut remains unclear; is it an innocent bystander in diarrhoeal disease caused by other organisms, or a pathogen causing subclinical to severe symptoms? Further investigation of the development of serological and local host response to C. difficile carriage may shed new light on disease mechanisms. Work is underway on defining a framework for diagnosis and management of paediatric CDI.
Collapse
Affiliation(s)
- E A Lees
- University of Liverpool Institute of Translational Medicine, Wolfson Centre, Block A: Waterhouse Building, 1-5 Brownlow Street, Liverpool, L69 3GL, UK.
| | - F Miyajima
- University of Liverpool Institute of Translational Medicine, Wolfson Centre, Block A: Waterhouse Building, 1-5 Brownlow Street, Liverpool, L69 3GL, UK
| | - M Pirmohamed
- University of Liverpool Institute of Translational Medicine, Wolfson Centre, Block A: Waterhouse Building, 1-5 Brownlow Street, Liverpool, L69 3GL, UK
| | - E D Carrol
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, Ronald Ross Building, West Derby Street, Liverpool, L69 7BE, UK
| |
Collapse
|
109
|
Abstract
BACKGROUND Clostridium difficile infection (CDI) affects patients with inflammatory bowel disease (IBD). The aim of this study was to compare humoral response to C. difficile toxins in IBD patients and control outpatients. METHODS We prospectively followed adult IBD patients and control subjects with serum and stool samples obtained at enrollment and during periods of CDI and tested by PCR. Semiquantitative serum levels of IgM, IgG, and IgA to C. difficile toxins A and B were measured. RESULTS Overall, 119 stool and 117 serum samples were obtained from 150 subjects. Different levels of IgA to toxin A (P = 0.0016) and toxin B (P = 0.0468) were noted between different IBD groups. Toxin A IgA levels were higher in the Crohn's disease group (P = 0.0321) and ileal pouch anal anastomosis (IPAA) group (P = 0.001) compared with the ulcerative colitis (UC) group, and toxin B IgA levels were higher in the IPAA group compared with the UC group (P = 0.0309). There were lower levels of toxin A IgA in IBD patients compared with those in subjects without new CDI (P = 0.0488) and higher levels in IBD patients with compared with those in subjects without CDI history before enrollment (P = 0.016). There were nonsignificant lower toxin A IgG levels in IBD patients compared with those in subjects without prior CDI (P = 0.095) and higher levels in control subjects with a history of CDI compared with IBD patients with prior CDI (P = 0.049). CONCLUSIONS Patients with UC have lower IgA levels to C. difficile toxins compared with those with Crohn's disease and those after IPAA. Patients with IBD with prior CDI failed to demonstrate any increase in antitoxin IgG. Our findings suggest that IBD patients may benefit from immunization strategies targeting C. difficile toxins.
Collapse
|
110
|
Sensitive assays enable detection of serum IgG antibodies against Clostridium difficile toxin A and toxin B in healthy subjects and patients with Clostridium difficile infection. Bioanalysis 2016; 8:611-23. [PMID: 26964649 DOI: 10.4155/bio-2015-0033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Pathogenic Clostridium difficile produces two proinflammatory exotoxins, toxin A and toxin B. Low level of serum antitoxin IgG antibodies is a risk factor for the development of primary and recurrent C. difficile infection (CDI). RESULTS We developed and validated two sensitive, titer-based electrochemiluminescence assays for the detection of serum antibody levels against C. difficile toxins A and B. These assays demonstrated excellent precision. The sensitivity of the assays allowed the detection of antitoxin A and antitoxin B IgG antibodies in all tested serum samples during assay validation. CONCLUSION The validated titer-based assays enable assessment of antitoxin A and antitoxin B IgG antibodies as potential biomarkers to identify patients with CDI at increased risk for CDI recurrence.
Collapse
|
111
|
Li YT, Cai HF, Wang ZH, Xu J, Fang JY. Systematic review with meta-analysis: long-term outcomes of faecal microbiota transplantation for Clostridium difficile infection. Aliment Pharmacol Ther 2016; 43:445-57. [PMID: 26662643 DOI: 10.1111/apt.13492] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 08/30/2015] [Accepted: 11/12/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Clostridium difficile infection is a major cause of nosocomial diarrhoea. AIM To evaluate long-term (≥90 days) efficacy and safety of faecal microbiota transplantation for C. difficile infection and explore the factors affecting the faecal microbiota transplantation outcomes. METHODS MEDLINE, the Cochrane Library and EMBASE were searched and only observational studies that utilised faecal microbiota transplantation for C. difficile infection with long-term follow-up duration (≥90 days) were included. Primary cure rate, overall recurrence rate and early (<90 days) and late (≥90 days) recurrence rate were calculated. RESULTS Eighteen observational studies with 611 patients were included. The primary cure rate was 91.2% (95% confidence interval, CI 86.7-94.8%). The overall recurrence rate was 5.5% (95% CI 2.2-10.3%). The early recurrence rate and late recurrence rate were 2.7% (95% CI 0.7-6.0%) and 1.7% (95% CI 0.4-4.2%) respectively. Most adverse events were expected, short-lived, self-limited and manageable. The association between faecal microbiota transplantation therapy and adverse events such as inflammatory bowel disease flare, infectious disease and autoimmune disease was a concern but remained insignificant. Old age (≥65 years) was identified as a risk factor for after faecal microbiota transplantation therapy. Upper gastrointestinal administration also results in less frequent primary cure. CONCLUSIONS Faecal microbiota transplantation seems to be a highly effective and robust therapy for recurrent C. difficile infection. However, more quality studies, such as randomised controlled trials and cohort studies with control groups, are needed to confirm its long-term efficacy and safety.
Collapse
Affiliation(s)
- Y-T Li
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Institute of Digestive Disease, Shanghai Jiao-Tong University School of Medicine, Shanghai, China.,Key Laboratory of Gastroenterology & Hepatology, Ministry of Health, Shanghai, China.,State Key Laboratory for Oncogenes and Related Genes, Shanghai, China
| | - H-F Cai
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Institute of Digestive Disease, Shanghai Jiao-Tong University School of Medicine, Shanghai, China.,Key Laboratory of Gastroenterology & Hepatology, Ministry of Health, Shanghai, China.,State Key Laboratory for Oncogenes and Related Genes, Shanghai, China
| | - Z-H Wang
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Institute of Digestive Disease, Shanghai Jiao-Tong University School of Medicine, Shanghai, China.,Key Laboratory of Gastroenterology & Hepatology, Ministry of Health, Shanghai, China.,State Key Laboratory for Oncogenes and Related Genes, Shanghai, China
| | - J Xu
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Institute of Digestive Disease, Shanghai Jiao-Tong University School of Medicine, Shanghai, China.,Key Laboratory of Gastroenterology & Hepatology, Ministry of Health, Shanghai, China.,State Key Laboratory for Oncogenes and Related Genes, Shanghai, China
| | - J-Y Fang
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Institute of Digestive Disease, Shanghai Jiao-Tong University School of Medicine, Shanghai, China.,Key Laboratory of Gastroenterology & Hepatology, Ministry of Health, Shanghai, China.,State Key Laboratory for Oncogenes and Related Genes, Shanghai, China
| |
Collapse
|
112
|
Almeida R, Gerbaba T, Petrof EO. Recurrent Clostridium difficile infection and the microbiome. J Gastroenterol 2016; 51:1-10. [PMID: 26153514 DOI: 10.1007/s00535-015-1099-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 06/16/2015] [Indexed: 02/04/2023]
Abstract
The diverse and densely populated gastrointestinal microbiota is essential for the regulation of host physiology and immune function. As our knowledge of the composition and function of the intestinal microbiota continues to expand, there is new interest in using these developments to tailor fecal microbiota transplantation (FMT) and microbial ecosystem therapeutics (MET) for a variety of diseases. The potential role of FMT and MET in the treatment of Clostridium difficile infection (CDI)-currently the leading nosocomial gastrointestinal infection-has proven highly effective for recurrent CDI, and has emerged as a paradigm shift in the treatment of this disease. The current review will serve as a summary of the key aspects of CDI, and will introduce the essential framework and challenges of FMT, as is currently practiced. MET represents the progression of conventional bacteriotherapy that fundamentally capitalizes on the restorative properties of intestinal bacterial communities and may be viewed as the culmination of a rationally designed therapeutic modality. As our understanding of the composition and function of the intestinal microbiota evolves, it will likely drive next-generation microbiota therapies for a range of medical conditions, such as inflammatory bowel disease, obesity, and metabolic syndrome.
Collapse
Affiliation(s)
- Rowena Almeida
- Gastrointestinal Diseases Research Unit, Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Teklu Gerbaba
- Gastrointestinal Diseases Research Unit, Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Elaine O Petrof
- Gastrointestinal Diseases Research Unit, Department of Medicine, Queen's University, Kingston, ON, Canada. .,Department of Medicine/ Division of Infectious Diseases, Queen's University, 76 Stuart Street, GIDRU Wing, Kingston, ON, K7L 2V7, Canada.
| |
Collapse
|
113
|
Abstract
INTRODUCTION Clostridium difficile infections are a leading cause of healthcare facility outbreaks of gastrointestinal illness that may have serious complications and a high rate of recurrent disease. Despite the availability of standard antibiotic treatments, data from national surveillance programs indicate that the incidence of this disease continues to increase, placing a heavy burden on healthcare systems. New emerging strategies are being tested to replace or augment these standard antibiotics. AREAS COVERED Thirty-two current investigational agents focusing on different strategies for both prevention and treatment of C. difficile infections are reviewed. Data was gathered from a literature search of public databases for published trials from 1999-November 13, 2015 and from the author's compendium of knowledge. Agents reviewed included 13 antibiotics, two antibiotic inactivators, seven bacteria or yeasts acting to enhance the normal microbiome, seven immunizing agents and three toxin binders. Of the 32 investigational treatments reviewed, 8 (25%) showed significant efficacy in phase II or III clinical trials and are actively being developed as new therapies for C. difficile infections. EXPERT OPINION A number of potential treatments have floundered during their development process, while others have shown promising results. The strongest efficacy has been in the areas of newer antibiotics, probiotics, monoclonal antibodies and vaccines. By targeting the pathogenic pathway of C. difficile infections, multiple strategies for prevention and treatment have been developed.
Collapse
Affiliation(s)
- Lynne V McFarland
- a Dept of Medicinal Chemistry , University of Washington , Seattle , WA , USA
| |
Collapse
|
114
|
Devera TS, Lang GA, Lanis JM, Rampuria P, Gilmore CL, James JA, Ballard JD, Lang ML. Memory B Cells Encode Neutralizing Antibody Specific for Toxin B from the Clostridium difficile Strains VPI 10463 and NAP1/BI/027 but with Superior Neutralization of VPI 10463 Toxin B. Infect Immun 2016; 84:194-204. [PMID: 26502913 PMCID: PMC4693989 DOI: 10.1128/iai.00011-15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 10/19/2015] [Indexed: 02/07/2023] Open
Abstract
Secreted toxin B (TcdB) substantially contributes to the pathology observed during Clostridium difficile infection. To be successfully incorporated into a vaccine, TcdB-based immunogens must stimulate the production of neutralizing antibody (Ab)-encoding memory B cells (Bmem cells). Despite numerous investigations, a clear analysis of Bmem cellular responses following vaccination against TcdB is lacking. B6 mice were therefore used to test the ability of a nontoxigenic C-terminal domain (CTD) fragment of TcdB to induce Bmem cells that encode TcdB-neutralizing antibody. CTD was produced from the historical VPI 10463 strain (CTD1) and from the hypervirulent strain NAP1/BI/027 (CTD2). It was then demonstrated that CTD1 induced strong recall IgG antibody titers, and this led to the development of functional Bmem cells that could be adoptively transferred to naive recipients. Bmem cell-driven neutralizing Ab responses conferred protection against lethal challenge with TcdB1. Further experiments revealed that an experimental adjuvant (Imject) and a clinical adjuvant (Alhydrogel) were compatible with Bmem cell induction. Reactivity of human Bmem cells to CTD1 was also evident in human peripheral blood mononuclear cells (PBMCs), suggesting that CTD1 could be a good vaccine immunogen. However, CTD2 induced strong Bmem cell-driven antibody titers, and the CTD2 antibody was neutralizing in vitro, but its protection against lethal challenge with TcdB2 was limited to delaying time to death. Therefore, CTD from different C. difficile strains may be a good immunogen for stimulating B cell memory that encodes in vitro neutralizing Ab but may be limited by variable protection against intoxication in vivo.
Collapse
Affiliation(s)
- T Scott Devera
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Gillian A Lang
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Jordi M Lanis
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Pragya Rampuria
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Casey L Gilmore
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Judith A James
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA Oklahoma Clinical and Translational Science Institute, Oklahoma City, Oklahoma, USA
| | - Jimmy D Ballard
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Mark L Lang
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
115
|
Schenck LP, Beck PL, MacDonald JA. Gastrointestinal dysbiosis and the use of fecal microbial transplantation in Clostridium difficile infection. World J Gastrointest Pathophysiol 2015; 6:169-180. [PMID: 26600975 PMCID: PMC4644881 DOI: 10.4291/wjgp.v6.i4.169] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/28/2015] [Accepted: 10/13/2015] [Indexed: 02/06/2023] Open
Abstract
The impact of antibiotics on the human gut microbiota is a significant concern. Antibiotic-associated diarrhea has been on the rise for the past few decades with the increasing usage of antibiotics. Clostridium difficile infections (CDI) have become one of the most prominent types of infectious diarrheal disease, with dramatically increased incidence in both the hospital and community setting worldwide. Studies show that variability in the innate host response may in part impact upon CDI severity in patients. That being said, CDI is a disease that shows the most prominent links to alterations to the gut microbiota, in both cause and treatment. With recurrence rates still relatively high, it is important to explore alternative therapies to CDI. Fecal microbiota transplantation (FMT) and other types of bacteriotherapy have become exciting avenues of treatment for CDI. Recent clinical trials have generated excitement for the use of FMT as a therapeutic option for CDI; however, the exact components of the human gut microbiota needed for protection against CDI have remained elusive. Additional investigations on the effects of antibiotics on the human gut microbiota and subsequent CDI will help reduce the socioeconomic burden of CDI and potentially lead to new therapeutic modalities.
Collapse
|
116
|
Zhao S, Ghose-Paul C, Zhang K, Tzipori S, Sun X. Immune-based treatment and prevention of Clostridium difficile infection. Hum Vaccin Immunother 2015; 10:3522-30. [PMID: 25668664 DOI: 10.4161/21645515.2014.980193] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Clostridium difficile (C. difficile) causes over 500,000 infections per year in the US, with an estimated 15,000 deaths and an estimated cost of $1-3 billion. Moreover, a continual rise in the incidence of severe C. difficile infection (CDI) has been observed worldwide. Currently, standard treatment for CDI is the administration of antibiotics. While effective, these treatments do not prevent and may contribute to a disease recurrence rate of 15-35%. Prevention of recurrence is one of the most challenging aspects in the field. A better knowledge of the molecular mechanisms of the disease, the host immune response and identification of key virulence factors of C. difficilenow permits the development of immune-based therapies. Antibodies specific for C. difficile toxins have been shown to effectively treat CDI and prevent disease relapse in animal models and in humans. Vaccination has been recognized as the most cost-effective treatment/prevention for CDI. This review will summarize CDI transmission, epidemiology, major virulent factors and highlights the rational and the development of immune-based approaches against this remerging threat.
Collapse
Key Words
- AAD, antibiotic-associated diarrhea
- CDI, Clostridium difficile infection
- CPD, cysteine proteinase domain
- GTD, glucosyltransferase domain
- HuMabs, human monoclonal antibodies
- IVIG, intravenous immunoglobulin
- RBD, receptor binding domain
- SLP, surface-layer protein
- TMD, transmembrane domain
- bacterial toxins
- clostridium difficile infection (CDI)
- immunotherapy
- mAb, monoclonal antibody
- monoclonal antibody
- vaccine
Collapse
Affiliation(s)
- Song Zhao
- a Department of Infectious Diseases and Global Health ; Tufts University Cummings School of Veterinary Medicine ; North Grafton , MA USA
| | | | | | | | | |
Collapse
|
117
|
Negm OH, Hamed MR, Dilnot EM, Shone CC, Marszalowska I, Lynch M, Loscher CE, Edwards LJ, Tighe PJ, Wilcox MH, Monaghan TM. Profiling Humoral Immune Responses to Clostridium difficile-Specific Antigens by Protein Microarray Analysis. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:1033-9. [PMID: 26178385 PMCID: PMC4550668 DOI: 10.1128/cvi.00190-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 07/02/2015] [Indexed: 01/05/2023]
Abstract
Clostridium difficile is an anaerobic, Gram-positive, and spore-forming bacterium that is the leading worldwide infective cause of hospital-acquired and antibiotic-associated diarrhea. Several studies have reported associations between humoral immunity and the clinical course of C. difficile infection (CDI). Host humoral immune responses are determined using conventional enzyme-linked immunosorbent assay (ELISA) techniques. Herein, we report the first use of a novel protein microarray assay to determine systemic IgG antibody responses against a panel of highly purified C. difficile-specific antigens, including native toxins A and B (TcdA and TcdB, respectively), recombinant fragments of toxins A and B (TxA4 and TxB4, respectively), ribotype-specific surface layer proteins (SLPs; 001, 002, 027), and control proteins (tetanus toxoid and Candida albicans). Microarrays were probed with sera from a total of 327 individuals with CDI, cystic fibrosis without diarrhea, and healthy controls. For all antigens, precision profiles demonstrated <10% coefficient of variation (CV). Significant correlation was observed between microarray and ELISA in the quantification of antitoxin A and antitoxin B IgG. These results indicate that microarray is a suitable assay for defining humoral immune responses to C. difficile protein antigens and may have potential advantages in throughput, convenience, and cost.
Collapse
Affiliation(s)
- Ola H Negm
- Immunology, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom Medical Microbiology and Immunology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed R Hamed
- Immunology, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom Medical Microbiology and Immunology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Elizabeth M Dilnot
- Immunology, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | | | - Mark Lynch
- Immunomodulation Research Group, Dublin City University, Dublin, Ireland
| | | | - Laura J Edwards
- Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Patrick J Tighe
- Immunology, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Mark H Wilcox
- Leeds Institute for Molecular Medicine, University of Leeds, Leeds, United Kingdom
| | - Tanya M Monaghan
- NIHR Nottingham Digestive Diseases Biomedical Research Unit, Nottingham, United Kingdom
| |
Collapse
|
118
|
Huang JH, Wu CW, Lien SP, Leng CH, Hsiao KN, Liu SJ, Chen HW, Siu LK, Chong P. Recombinant lipoprotein-based vaccine candidates against C. difficile infections. J Biomed Sci 2015; 22:65. [PMID: 26245825 PMCID: PMC4527207 DOI: 10.1186/s12929-015-0171-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 07/29/2015] [Indexed: 12/18/2022] Open
Abstract
Background Opportunistically nosocomial infections in hospitalized patients are often related to Clostridium difficile infections (CDI) due to disruption of the intestinal micro-flora by antibiotic therapies during hospitalization. Clostridial exotoxins A and B (TcdA and TcdB) specifically bind to unknown glycoprotein(s) in the host intestine, disrupt the intestinal barrier leading to acute inflammation and diarrhea. The C-terminal receptor binding domain of TcdA (A-rRBD) has been shown to elicit antibody responses that neutralize TcdA toxicity in Vero cell cytotoxicity assays, but not effectively protect hamsters against a lethal dose challenge of C. difficile spores. To develop an effective recombinant subunit vaccine against CDI, A-rRBD was lipidated (rlipoA-RBD) as a rational design to contain an intrinsic adjuvant, a toll-like receptor 2 agonist and expressed in Escherichia coli. Results The purified rlipoA-RBD was characterized immunologically and found to have the following properties: (a) mice, hamsters and rabbits vaccinated with 3 μg of rlipoA-RBD produced strong antibody responses that neutralized TcdA toxicity in Vero cell cytotoxicity assays; furthermore, the neutralization titer was comparable to those obtained from antisera immunized either with 10 μg of TcdA toxoid or 30 μg of A-rRBD; (b) rlipoA-RBD elicited immune responses and protected mice from TcdA challenge, but offered insignificant protection (10 to 20 %) against C. difficile spores challenge in hamster models; (c) only rlipoA-RBD formulated with B-rRBD consistently confers protection (90 to 100 %) in the hamster challenge model; and (d) rlipoA-RBD was found to be 10-fold more potent than A-rRBD as an adjuvant to enhancing immune responses against a poor antigen such as ovalbumin. Conclusion These results indicate that rlipoA-RBD formulated with B-rRBD could be an excellent vaccine candidate for preclinical studies and future clinical trials.
Collapse
Affiliation(s)
- Jui-Hsin Huang
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan. .,Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan.
| | - Chia-Wei Wu
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan.
| | - Shu-Pei Lien
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan.
| | - Chih-Hsiang Leng
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan.
| | - Kuang-Nan Hsiao
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan.
| | - Shih-Jen Liu
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan.
| | - Hsin-Wei Chen
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan.
| | - Leung-Kei Siu
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan.
| | - Pele Chong
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan. .,Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan. .,Graduate Institute of Immunology, China Medical University, Taichung, Taiwan.
| |
Collapse
|
119
|
Abstract
Colonization with toxigenic Clostridium difficile may be associated with a wide spectrum of clinical presentation ranging from asymptomatic carriage to mild diarrhea to life-threatening colitis. Over the last 15 years, there has been a marked increase in the incidence of C. difficile infection, which predominantly affects elderly patients on antibiotics. More recently, there has been significant interest in the association between inflammatory bowel disease (IBD) and C. difficile infection. This review article discusses in some detail current knowledge of the mechanisms by which C. difficile toxins may mediate mucosal inflammation, together with the role of cell wall components of the microorganism in disease pathogenesis. Innate and adaptive host responses to C. difficile toxins and other components are described and include consideration of the potential role of known mucosal changes in IBD that may lead to an enhanced inflammatory response in the presence of C. difficile infection. Recent studies, which have characterized resident microbiota that may mediate protection against colonization by C. difficile, including their mechanisms of action, are also discussed. This includes the role of bile acids and 7α-dehydroxylase-expressing bacteria, such as Clostridium scindens. Recent studies suggest a higher carriage rate of C. difficile in patients with IBD. It is anticipated that future studies will determine the role of dysbiosis in IBD in predisposing to colonization with C. difficile.
Collapse
|
120
|
Kachrimanidou M, Sarmourli T, Skoura L, Metallidis S, Malisiovas N. Clostridium difficile infection: New insights into therapeutic options. Crit Rev Microbiol 2015; 42:773-9. [PMID: 25955884 DOI: 10.3109/1040841x.2015.1027171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Clostridium difficile infection (CDI) is an important cause of mortality and morbidity in healthcare settings and represents a major social and economic burden. The major virulence determinants are large clostridial toxins, toxin A (TcdA) and toxin B (TcdB), encoded within the pathogenicity locus. Traditional therapies, such as metronidazole and vancomycin, frequently lead to a vicious circle of recurrences due to their action against normal human microbiome. New disease management strategies together with the development of novel therapeutic and containment approaches are needed in order to better control outbreaks and treat patients. This article provides an overview of currently available CDI treatment options and discusses the most promising therapies under development.
Collapse
Affiliation(s)
- Melina Kachrimanidou
- a Department of Microbiology , Medical School, Aristotle University of Thessaloniki , Greece , Thessaloniki , Greece and
| | - Theopisti Sarmourli
- a Department of Microbiology , Medical School, Aristotle University of Thessaloniki , Greece , Thessaloniki , Greece and
| | - Lemonia Skoura
- a Department of Microbiology , Medical School, Aristotle University of Thessaloniki , Greece , Thessaloniki , Greece and
| | - Symeon Metallidis
- b Infectious Diseases Division, Department of Internal Medicine , Medical School, Aristotle University of Thessaloniki , Thessaloniki , Greece
| | - Nikolaos Malisiovas
- a Department of Microbiology , Medical School, Aristotle University of Thessaloniki , Greece , Thessaloniki , Greece and
| |
Collapse
|
121
|
A Combination of Three Fully Human Toxin A- and Toxin B-Specific Monoclonal Antibodies Protects against Challenge with Highly Virulent Epidemic Strains of Clostridium difficile in the Hamster Model. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:711-25. [PMID: 25924765 DOI: 10.1128/cvi.00763-14] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 04/21/2015] [Indexed: 12/21/2022]
Abstract
Clostridium difficile infection (CDI) is the principal cause of nosocomial diarrhea and pseudomembranous colitis associated with antibiotic therapy. Recent increases in the number of outbreaks attributed to highly virulent antibiotic-resistant strains underscore the importance of identifying efficacious alternatives to antibiotics to control this infection. CDI is mediated by two large exotoxins, toxins A and B. Strong humoral toxin-specific immune responses are associated with recovery and a lack of disease recurrence, whereas insufficient humoral responses are associated with recurrent CDI. Multiple approaches targeting these toxins, including intravenous immunoglobulin, neutralizing polymers, active vaccines, and, most recently, monoclonal antibodies (MAbs), have been explored, with various degrees of success. In this study, we describe the characterization of the first MAbs isolated from healthy human donors using a high-throughput B-cell cloning strategy. The MAbs were selected based on their ability to inhibit the actions of toxins A and B in vitro and because of their in vivo efficacy in a hamster challenge model. A potent 2-MAb cocktail was identified and then further potentiated by the addition of a second anti-toxin B MAb. This 3-MAb combination protected animals against mortality and also reduced the severity and duration of diarrhea associated with challenge with highly virulent strains of C. difficile toxinotypes 0 and III. This highly efficacious cocktail consists of one MAb specific to the receptor binding domain of toxin A and two MAbs specific to nonoverlapping regions of the glucosyltransferase domain of toxin B. This MAb combination offers great potential as a nonantibiotic treatment for the prevention of recurrent CDI.
Collapse
|
122
|
Abstract
The best laboratory diagnostic approach to detect Clostridium difficile infection (CDI) is the subject of ongoing debate. In the United States, nucleic acid amplification tests (NAAT) have become the most widely used tests for making this diagnosis. Detection of toxin in stool may be a better predictor of CDI disease and severity. Laboratories that have switched from toxin-based to NAAT-based methods have significantly higher CDI detection rates. The important issue is whether all NAAT-positive patients have CDI or at least some of those patients are excretors of the organism and do not have clinical disease.
Collapse
Affiliation(s)
- Peter H Gilligan
- Clinical Microbiology-Immunology Laboratories, Microbiology-Immunology, UNC Health Care, UNC School of Medicine, CB 7600, Chapel Hill, NC 27516, USA; Pathology-Laboratory Medicine, UNC School of Medicine, CB 7600, Chapel Hill, NC 27516, USA.
| |
Collapse
|
123
|
Abstract
Clostridium difficile is a spore-forming anaerobic gram-positive organism that is the leading cause of antibiotic-associated nosocomial infectious diarrhea in the Western world. This article describes the evolving epidemiology of C difficile infection (CDI) in the twenty-first century, evaluates the importance of vaccines against the disease, and defines the roles of both innate and adaptive host immune responses in CDI. The effects of passive immunotherapy and active vaccination against CDI in both humans and animals are also discussed.
Collapse
Affiliation(s)
- Chandrabali Ghose
- Aaron Diamond AIDS Research Center, 455 First Avenue, 7th Floor, New York, NY 10016, USA.
| | - Ciarán P Kelly
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| |
Collapse
|
124
|
Abstract
IMPORTANCE Since 2000, the incidence and severity of Clostridium difficile infection (CDI) have increased. OBJECTIVE To review current evidence regarding best practices for the diagnosis and treatment of CDI in adults (age ≥ 18 years). EVIDENCE REVIEW Ovid MEDLINE and Cochrane databases were searched using keywords relevant to the diagnosis and treatment of CDI in adults. Articles published between January 1978 and October 31, 2014, were selected for inclusion based on targeted keyword searches, manual review of bibliographies, and whether the article was a guideline, systematic review, or meta-analysis published within the past 10 years. Of 4682 articles initially identified, 196 were selected for full review. Of these, the most pertinent 116 articles were included. Clinical trials, large observational studies, and more recently published articles were prioritized in the selection process. FINDINGS Laboratory testing cannot distinguish between asymptomatic colonization and symptomatic infection with C difficile. Diagnostic approaches are complex due to the availability of multiple testing strategies. Multistep algorithms using polymerase chain reaction (PCR) for the toxin gene(s) or single-step PCR on liquid stool samples have the best test performance characteristics (for multistep: sensitivity was 0.68-1.00 and specificity was 0.92-1.00; and for single step: sensitivity was 0.86-0.92 and specificity was 0.94-0.97). Vancomycin and metronidazole are first-line therapies for most patients, although treatment failures have been associated with metronidazole in severe or complicated cases of CDI. Recent data demonstrate clinical success rates of 66.3% for metronidazole vs 78.5% for vancomycin for severe CDI. Newer therapies show promising results, including fidaxomicin (similar clinical cure rates to vancomycin, with lower recurrence rates for fidaxomicin, 15.4% vs vancomycin, 25.3%; P = .005) and fecal microbiota transplantation (response rates of 83%-94% for recurrent CDI). CONCLUSIONS AND RELEVANCE Diagnostic testing for CDI should be performed only in symptomatic patients. Treatment strategies should be based on disease severity, history of prior CDI, and the individual patient's risk of recurrence. Vancomycin is the treatment of choice for severe or complicated CDI, with or without adjunctive therapies. Metronidazole is appropriate for mild disease. Fidaxomicin is a therapeutic option for patients with recurrent CDI or a high risk of recurrence. Fecal microbiota transplantation is associated with symptom resolution of recurrent CDI but its role in primary and severe CDI is not established.
Collapse
Affiliation(s)
- Natasha Bagdasarian
- Division of Infectious Disease and Department of Infection Control, St John Hospital and Medical Center, Detroit, Michigan
- Wayne State University, Department of Internal Medicine, Detroit, Michigan
| | - Krishna Rao
- University of Michigan Medical School, Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Health System, Ann Arbor, Michigan
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Preeti N. Malani
- University of Michigan Medical School, Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Health System, Ann Arbor, Michigan
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| |
Collapse
|
125
|
Abstract
Clostridium difficile is associated with a spectrum of clinical manifestations ranging from asymptomatic carriage to severe life-threatening pseudomembranous colitis. Current perspectives indicate that C difficile pathogenesis is a multifactorial disease process dictated by pathogenic toxin production, gut microbial dysbiosis, and altered host inflammatory responses. This article summarizes recent findings underpinning the cellular and molecular mechanisms regulating bacterial virulence and sheds new light on the critical roles of the host immune response, intestinal microbiota, and metabolome in mediating disease pathogenesis.
Collapse
Affiliation(s)
- Tanya M Monaghan
- Biomedical Research Unit, NIHR Nottingham Digestive Diseases Centre, Nottingham University Hospitals NHS Trust, Derby Road, Nottingham NG7 2UH, UK.
| |
Collapse
|
126
|
Mechanisms of protection against Clostridium difficile infection by the monoclonal antitoxin antibodies actoxumab and bezlotoxumab. Infect Immun 2014; 83:822-31. [PMID: 25486992 DOI: 10.1128/iai.02897-14] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Clostridium difficile infection (CDI) represents the most prevalent cause of antibiotic-associated gastrointestinal infections in health care facilities in the developed world. Disease symptoms are caused by the two homologous exotoxins, TcdA and TcdB. Standard therapy for CDI involves administration of antibiotics that are associated with a high rate of disease recurrence, highlighting the need for novel treatment paradigms that target the toxins rather than the organism itself. A combination of human monoclonal antibodies, actoxumab and bezlotoxumab, directed against TcdA and TcdB, respectively, has been shown to decrease the rate of recurrence in patients treated with standard-of-care antibiotics. However, the exact mechanism of antibody-mediated protection is poorly understood. In this study, we show that the antitoxin antibodies are protective in multiple murine models of CDI, including systemic and local (gut) toxin challenge models, as well as primary and recurrent models of infection in mice. Systemically administered actoxumab-bezlotoxumab prevents both the damage to the gut wall and the inflammatory response, which are associated with C. difficile in these models, including in mice challenged with a strain of the hypervirulent ribotype 027. Furthermore, mutant antibodies (N297Q) that do not bind to Fcγ receptors provide a level of protection similar to that of wild-type antibodies, demonstrating that the mechanism of protection is through direct neutralization of the toxins and does not involve host effector functions. These data provide a mechanistic basis for the prevention of recurrent disease observed in CDI patients in clinical trials.
Collapse
|
127
|
Bauer MP, Nibbering PH, Poxton IR, Kuijper EJ, van Dissel JT. Humoral immune response as predictor of recurrence in Clostridium difficile infection. Clin Microbiol Infect 2014; 20:1323-8. [PMID: 25041274 DOI: 10.1111/1469-0691.12769] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 04/30/2014] [Accepted: 07/09/2014] [Indexed: 11/26/2022]
Abstract
Low serum concentrations of antibodies directed against the toxins TcdA and TcdB have been associated with a higher risk of recurrence of Clostridium difficile infection (CDI) after successful antibiotic treatment. However, there are conflicting reports. Herein, we compared serum levels of antibodies of patients with a single episode of CDI with those of patients who subsequently suffered a recurrence. We used a serum bank from patients who received an experimental whey protein product following successful antibiotic treatment for CDI. We determined levels of IgA and IgG directed against TcdA, TcdB and non-toxin cell surface antigens in serum collected directly and 3 weeks after completing a 10-day course of antibiotic treatment for CDI. We also developed an objective flow cytometry-based assay to determine the proportion of cells exhibiting cytopathic effect after exposure to TcdB. Using this method, we measured the TcdB-neutralizing capacity of sera. We compared the results for patients without a subsequent recurrence with those of patients who suffered a recurrence within 60 days after completing the antibiotic treatment. Advanced age, comorbidity other than immunocompromised state and low serum levels of anti-TcdA and anti-TcdB antibodies were associated with recurrence, whereas serum levels of antibodies directed against cell surface antigens were not. Serum TcdB-neutralizing capacity, which correlated only weakly with serum IgG anti-TcdB, was not significantly associated with recurrence.
Collapse
Affiliation(s)
- M P Bauer
- Department of Infectious Diseases, Centre for Infectious Diseases, Leiden University Medical Centre, Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
128
|
Toxin-mediated paracellular transport of antitoxin antibodies facilitates protection against Clostridium difficile infection. Infect Immun 2014; 83:405-16. [PMID: 25385797 DOI: 10.1128/iai.02550-14] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The exotoxins TcdA and TcdB are the major virulence factors of Clostridium difficile. Circulating neutralizing antitoxin antibodies are protective in C. difficile infection (CDI), as demonstrated, in part, by the protective effects of actoxumab and bezlotoxumab, which bind to and neutralize TcdA and TcdB, respectively. The question of how systemic IgG antibodies neutralize toxins in the gut lumen remains unresolved, although it has been suggested that the Fc receptor FcRn may be involved in active antibody transport across the gut epithelium. In this study, we demonstrated that genetic ablation of FcRn and excess irrelevant human IgG have no impact on actoxumab-bezlotoxumab-mediated protection in murine and hamster models of CDI, suggesting that Fc-dependent transport of antibodies across the gut wall is not required for efficacy. Tissue distribution studies in hamsters suggest, rather, that the transport of antibodies depends on toxin-induced damage to the gut lining. In an in vitro two-dimensional culture system that mimics the architecture of the intestinal mucosal epithelium, toxins on the apical side of epithelial cell monolayers are neutralized by basolateral antibodies, and antibody transport across the cell layer is dramatically increased upon addition of toxin to the apical side. Similar data were obtained with F(ab')2 fragments, which lack an Fc domain, consistent with FcRn-independent paracellular, rather than transcellular, transport of antibodies. Kinetic studies show that initial damage caused by apical toxin is required for efficient neutralization by basolateral antibodies. These data may represent a general mechanism of humoral response-mediated protection against enteric pathogens.
Collapse
|
129
|
Cohen OR, Steele JA, Zhang Q, Schmidt DJ, Wang Y, Hamel PES, Beamer G, Xu B, Tzipori S. Systemically administered IgG anti-toxin antibodies protect the colonic mucosa during infection with Clostridium difficile in the piglet model. PLoS One 2014; 9:e111075. [PMID: 25347821 PMCID: PMC4210241 DOI: 10.1371/journal.pone.0111075] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 09/24/2014] [Indexed: 01/11/2023] Open
Abstract
The use of anti-toxin human monoclonal antibodies (HMab) as treatment for C. difficile infection has been investigated in animal models and human clinical trials as an alternative to or in combination with traditional antibiotic therapy. While HMab therapy appears to be a promising option, how systemically administered IgG antibodies protect the colonic mucosa during Clostridium difficile infection is unknown. Using the gnotobiotic piglet model of Clostridium difficile infection, we administered a mixture of anti-TcdA and anti-TcdB HMabs systemically to piglets infected with either pathogenic or non-pathogenic C. difficile strains. The HMabs were present throughout the small and large intestinal tissue of both groups, but significant HMabs were present in the lumen of the large intestines only in the pathogenic strain-infected group. Similarly, HMabs measured in the large intestine over a period of 2-4 days following antibody administration were not significantly different over time in the gut mucosa among the groups, but concentrations in the lumen of the large intestine were again consistently higher in the pathogenic strain-infected group. These results indicate that systemically administered HMab IgG reaches the gut mucosa during the course of CDI, protecting the host against systemic intoxication, and that leakage through the damaged colon likely protects the mucosa from further damage, allowing initiation of repair and recovery.
Collapse
Affiliation(s)
- Ocean R. Cohen
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Jennifer A. Steele
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Quanshun Zhang
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Diane J. Schmidt
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Yuankai Wang
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Philip E. S. Hamel
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Gillian Beamer
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Bingling Xu
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
130
|
Sun X, Hirota SA. The roles of host and pathogen factors and the innate immune response in the pathogenesis of Clostridium difficile infection. Mol Immunol 2014; 63:193-202. [PMID: 25242213 DOI: 10.1016/j.molimm.2014.09.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/03/2014] [Accepted: 09/03/2014] [Indexed: 02/08/2023]
Abstract
Clostridium difficile (C. difficile) is the most common cause of nosocomial antibiotic-associated diarrhea and the etiologic agent of pseudomembranous colitis. The clinical manifestation of C. difficile infection (CDI) is highly variable, from asymptomatic carriage, to mild self-limiting diarrhea, to the more severe pseudomembranous colitis. Furthermore, in extreme cases, colonic inflammation and tissue damage can lead to toxic megacolon, a condition requiring surgical intervention. C. difficile expresses two key virulence factors; the exotoxins, toxin A (TcdA) and toxin B (TcdB), which are glucosyltransferases that target host-cell monomeric GTPases. In addition, some hypervirulent strains produce a third toxin, binary toxin or C. difficile transferase (CDT), which may contribute to the pathogenesis of CDI. More recently, other factors such as surface layer proteins (SLPs) and flagellin have also been linked to the inflammatory responses observed in CDI. Although the adaptive immune response can influence the severity of CDI, the innate immune responses to C. difficile and its toxins play crucial roles in CDI onset, progression, and overall prognosis. Despite this, the innate immune responses in CDI have drawn relatively little attention from clinical researchers. Targeting these responses may prove useful clinically as adjuvant therapies, especially in refractory and/or recurrent CDI. This review will focus on recent advances in our understanding of how C. difficile and its toxins modulate innate immune responses that contribute to CDI pathogenesis.
Collapse
Affiliation(s)
- Xingmin Sun
- Tufts University Cummings School of Veterinary Medicine, Department of Infectious Diseases and Global Health, North Grafton, MA 01536, USA; Tufts University, Clinical and Translational Science Institute, Boston, MA 02111, USA.
| | - Simon A Hirota
- University of Calgary, Snyder Institute for Chronic Diseases, Departments of Physiology & Pharmacology and Microbiology, Immunology & Infectious Diseases, Calgary, AB T2N4N1, Canada
| |
Collapse
|
131
|
Clostridium difficile-induced colitis in mice is independent of leukotrienes. Anaerobe 2014; 30:90-8. [PMID: 25230329 DOI: 10.1016/j.anaerobe.2014.09.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 09/04/2014] [Accepted: 09/07/2014] [Indexed: 02/03/2023]
Abstract
Clostridium difficile is the major cause of antibiotic-associated diarrhea and pseudomembranous colitis in healthcare settings. However, the host factors involved in the intestinal inflammatory response and pathogenesis of C. difficile infection (CDI) are largely unknown. Here we investigated the role of leukotrienes (LTs), a group of pro-inflammatory lipid mediators, in CDI. Notably, the neutrophil chemoattractant LTB4, but not cysteinyl (cys) LTs, was induced in the intestine of C57BL/6 mice infected with either C. difficile strain VPI 10463 or strain 630. Genetic or pharmacological ablation of LT production did not ameliorate C. difficile colitis or clinical signs of disease in infected mice. Histological analysis demonstrated that intestinal neutrophilic inflammation, edema and tissue damage in mice during acute and severe CDI were not modulated in the absence of LTs. In addition, CDI induced a burst of cytokines in the intestine of infected mice in a LT-independent manner. Serum levels of anti-toxin A immunoglobulin (Ig) G levels were also not modulated by endogenous LTs. Collectively, our results do not support a role for LTs in modulating host susceptibility to CDI in mice.
Collapse
|
132
|
Debast SB, Bauer MP, Kuijper EJ. European Society of Clinical Microbiology and Infectious Diseases: update of the treatment guidance document for Clostridium difficile infection. Clin Microbiol Infect 2014; 20 Suppl 2:1-26. [PMID: 24118601 DOI: 10.1111/1469-0691.12418] [Citation(s) in RCA: 792] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 09/22/2013] [Accepted: 09/27/2013] [Indexed: 12/11/2022]
Abstract
In 2009 the first European Society of Clinical Microbiology and Infection (ESCMID) treatment guidance document for Clostridium difficile infection (CDI) was published. The guideline has been applied widely in clinical practice. In this document an update and review on the comparative effectiveness of the currently available treatment modalities of CDI is given, thereby providing evidence-based recommendations on this issue. A computerized literature search was carried out to investigate randomized and non-randomized trials investigating the effect of an intervention on the clinical outcome of CDI. The Grades of Recommendation Assessment, Development and Evaluation (GRADE) system was used to grade the strength of our recommendations and the quality of the evidence. The ESCMID and an international team of experts from 11 European countries supported the process. To improve clinical guidance in the treatment of CDI, recommendations are specified for various patient groups, e.g. initial non-severe disease, severe CDI, first recurrence or risk for recurrent disease, multiple recurrences and treatment of CDI when oral administration is not possible. Treatment options that are reviewed include: antibiotics, toxin-binding resins and polymers, immunotherapy, probiotics, and faecal or bacterial intestinal transplantation. Except for very mild CDI that is clearly induced by antibiotic usage antibiotic treatment is advised. The main antibiotics that are recommended are metronidazole, vancomycin and fidaxomicin. Faecal transplantation is strongly recommended for multiple recurrent CDI. In case of perforation of the colon and/or systemic inflammation and deteriorating clinical condition despite antibiotic therapy, total abdominal colectomy or diverting loop ileostomy combined with colonic lavage is recommended.
Collapse
|
133
|
Gupta SB, Dubberke ER. Overview and changing epidemiology of Clostridium difficile infection. SEMINARS IN COLON AND RECTAL SURGERY 2014. [DOI: 10.1053/j.scrs.2014.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
134
|
Heinrichs JH, Therien AG. Prevention of Clostridium difficile infections—The role of vaccines and therapeutic immunoglobulins. SEMINARS IN COLON AND RECTAL SURGERY 2014. [DOI: 10.1053/j.scrs.2014.05.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
135
|
Abstract
Clostridium difficile infection (CDI) is the most common infectious cause of healthcare-acquired diarrhoea. Outcomes of C. difficile colonization are varied, from asymptomatic carriage to fulminant colitis and death, due in part to the interplay between the pathogenic virulence factors of the bacterium and the counteractive immune responses of the host. Secreted toxins A and B are the major virulence factors of C. difficile and induce a profound inflammatory response by intoxicating intestinal epithelial cells causing proinflammatory cytokine release. Host cell necrosis, vascular permeability and neutrophil infiltration lead to an elevated white cell count, profuse diarrhoea and in severe cases, dehydration, hypoalbuminaemia and toxic megacolon. Other bacterial virulence factors, including surface layer proteins and flagella proteins, are detected by host cell surface signal molecules that trigger downstream cell-mediated immune pathways. Human studies have identified a role for serum and faecal immunoglobulin levels in protection from disease, but the recent development of a mouse model of CDI has enabled studies into the precise molecular interactions that trigger the immune response during infection. Key effector molecules have been identified that can drive towards a protective anti-inflammatory response or a damaging proinflammatory response. The limitations of current antimicrobial therapies for CDI have led to the development of both active and passive immunotherapies, none of which have, as yet been formally approved for CDI. However, recent advances in our understanding of the molecular basis of host immune protection against CDI may provide an exciting opportunity for novel therapeutic developments in the future.
Collapse
Affiliation(s)
- Katie Solomon
- School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Republic of Ireland
| |
Collapse
|
136
|
An optimized, synthetic DNA vaccine encoding the toxin A and toxin B receptor binding domains of Clostridium difficile induces protective antibody responses in vivo. Infect Immun 2014; 82:4080-91. [PMID: 25024365 DOI: 10.1128/iai.01950-14] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Clostridium difficile-associated disease (CDAD) constitutes a large majority of nosocomial diarrhea cases in industrialized nations and is mediated by the effects of two secreted toxins, toxin A (TcdA) and toxin B (TcdB). Patients who develop strong antitoxin antibody responses can clear C. difficile infection and remain disease free. Key toxin-neutralizing epitopes have been found within the carboxy-terminal receptor binding domains (RBDs) of TcdA and TcdB, which has generated interest in developing the RBD as a viable vaccine target. While numerous platforms have been studied, very little data describes the potential of DNA vaccination against CDAD. Therefore, we created highly optimized plasmids encoding the RBDs from TcdA and TcdB in which any putative N-linked glycosylation sites were altered. Mice and nonhuman primates were immunized intramuscularly, followed by in vivo electroporation, and in these animal models, vaccination induced significant levels of both anti-RBD antibodies (blood and stool) and RBD-specific antibody-secreting cells. Further characterization revealed that sera from immunized mice and nonhuman primates could detect RBD protein from transfected cells, as well as neutralize purified toxins in an in vitro cytotoxicity assay. Mice that were immunized with plasmids or given nonhuman-primate sera were protected from a lethal challenge with purified TcdA and/or TcdB. Moreover, immunized mice were significantly protected when challenged with C. difficile spores from homologous (VPI 10463) and heterologous, epidemic (UK1) strains. These data demonstrate the robust immunogenicity and efficacy of a TcdA/B RBD-based DNA vaccine in preclinical models of acute toxin-associated and intragastric, spore-induced colonic disease.
Collapse
|
137
|
Kelly CR, Ihunnah C, Fischer M, Khoruts A, Surawicz C, Afzali A, Aroniadis O, Barto A, Borody T, Giovanelli A, Gordon S, Gluck M, Hohmann EL, Kao D, Kao JY, McQuillen DP, Mellow M, Rank KM, Rao K, Ray A, Schwartz MA, Singh N, Stollman N, Suskind DL, Vindigni SM, Youngster I, Brandt L. Fecal microbiota transplant for treatment of Clostridium difficile infection in immunocompromised patients. Am J Gastroenterol 2014; 109:1065-71. [PMID: 24890442 PMCID: PMC5537742 DOI: 10.1038/ajg.2014.133] [Citation(s) in RCA: 496] [Impact Index Per Article: 45.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 04/21/2014] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Patients who are immunocompromised (IC) are at increased risk of Clostridium difficile infection (CDI), which has increased to epidemic proportions over the past decade. Fecal microbiota transplantation (FMT) appears effective for the treatment of CDI, although there is concern that IC patients may be at increased risk of having adverse events (AEs) related to FMT. This study describes the multicenter experience of FMT in IC patients. METHODS A multicenter retrospective series was performed on the use of FMT in IC patients with CDI that was recurrent, refractory, or severe. We aimed to describe rates of CDI cure after FMT as well as AEs experienced by IC patients after FMT. A 32-item questionnaire soliciting demographic and pre- and post-FMT data was completed for 99 patients at 16 centers, of whom 80 were eligible for inclusion. Outcomes included (i) rates of CDI cure after FMT, (ii) serious adverse events (SAEs) such as death or hospitalization within 12 weeks of FMT, (iii) infection within 12 weeks of FMT, and (iv) AEs (related and unrelated) to FMT. RESULTS Cases included adult (75) and pediatric (5) patients treated with FMT for recurrent (55%), refractory (11%), and severe and/or overlap of recurrent/refractory and severe CDI (34%). In all, 79% were outpatients at the time of FMT. The mean follow-up period between FMT and data collection was 11 months (range 3-46 months). Reasons for IC included: HIV/AIDS (3), solid organ transplant (19), oncologic condition (7), immunosuppressive therapy for inflammatory bowel disease (IBD; 36), and other medical conditions/medications (15). The CDI cure rate after a single FMT was 78%, with 62 patients suffering no recurrence at least 12 weeks post FMT. Twelve patients underwent repeat FMT, of whom eight had no further CDI. Thus, the overall cure rate was 89%. Twelve (15%) had any SAE within 12 weeks post FMT, of which 10 were hospitalizations. Two deaths occurred within 12 weeks of FMT, one of which was the result of aspiration during sedation for FMT administered via colonoscopy; the other was unrelated to FMT. None suffered infections definitely related to FMT, but two patients developed unrelated infections and five had self-limited diarrheal illness in which no causal organism was identified. One patient had a superficial mucosal tear caused by the colonoscopy performed for the FMT, and three patients reported mild, self-limited abdominal discomfort post FMT. Five (14% of IBD patients) experienced disease flare post FMT. Three ulcerative colitis (UC) patients underwent colectomy related to course of UC >100 days after FMT. CONCLUSIONS This series demonstrates the effective use of FMT for CDI in IC patients with few SAEs or related AEs. Importantly, there were no related infectious complications in these high-risk patients.
Collapse
Affiliation(s)
- Colleen R. Kelly
- Division of Gastroenterology, Brown Alpert Medical School, Women’s Medicine Collaborative, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Chioma Ihunnah
- Division of Gastroenterology, Brown Alpert Medical School, Women’s Medicine Collaborative, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Monika Fischer
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | - Anita Afzali
- University of Washington School of Medicine, Seattle, Washington, USA
| | - Olga Aroniadis
- Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Amy Barto
- Lahey Clinic Hospital and Medical Center, Tufts University School of Medicine, Burlington, Massachusetts, USA
| | - Thomas Borody
- Centre for Digestive Diseases, Five Dock, Sydney, New South Wales, Australia
| | - Andrea Giovanelli
- Northern California Gastroenterology Consultants, Inc., Oakland, California, USA
| | - Shelley Gordon
- California Pacific Medical Center, San Francisco, California, USA
| | - Michael Gluck
- Virginia Mason Medical Center, Seattle, Washington, USA
| | - Elizabeth L. Hohmann
- Massachusetts General Hospital and Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dina Kao
- University of Alberta, Edmonton, Alberta, Canada
| | - John Y. Kao
- University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel P. McQuillen
- Lahey Clinic Hospital and Medical Center, Tufts University School of Medicine, Burlington, Massachusetts, USA
| | - Mark Mellow
- Integris Baptist Medical Center, Oklahoma, Oklahoma, USA
| | - Kevin M. Rank
- University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Krishna Rao
- University of Michigan, Ann Arbor, Michigan, USA
| | - Arnab Ray
- Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | | | - Namita Singh
- Seattle Children’s Hospital, Seattle, Washington, USA
| | - Neil Stollman
- Northern California Gastroenterology Consultants, Inc., Oakland, California, USA
| | | | | | - Ilan Youngster
- Massachusetts General Hospital and Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lawrence Brandt
- Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
138
|
Petrof EO, Khoruts A. From stool transplants to next-generation microbiota therapeutics. Gastroenterology 2014; 146:1573-1582. [PMID: 24412527 PMCID: PMC4221437 DOI: 10.1053/j.gastro.2014.01.004] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 12/23/2013] [Accepted: 01/06/2014] [Indexed: 12/19/2022]
Abstract
The epidemic of Clostridium difficile infection fueled by new virulent strains of the organism has led to increased use of fecal microbiota transplantation (FMT). The procedure is effective for even the most desperate cases after failure of multiple courses of antibiotics. The approach recognizes microbiota to be integral to normal human physiology, and microbiota being used in FMT represents a new class of therapeutics. Imbalance in the composition and altered activity of the microbiota are associated with many diseases. Consequently, there is growing interest in applying FMT to non-C difficile indications. However, this may succeed only if microbiota therapeutics are developed systematically, based on mechanistic understanding, and applying up-to-date principles of microbial ecology. We discuss 2 pathways in the development of this new therapeutic class: whole microbial communities separated from donor stool and an assembly of specific fecal microorganisms grown in vitro.
Collapse
Affiliation(s)
- Elaine O. Petrof
- Department of Medicine, Division of Infectious Diseases & Gastrointestinal Research Unit; Queens University and Kingston General Hospital, Kingston, ON, Canada
| | - Alexander Khoruts
- Center for Immunology and Department of Medicine, Division of Gastroenterology; University of Minnesota, MN, USA
| |
Collapse
|
139
|
Antibodies for treatment of Clostridium difficile infection. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:913-23. [PMID: 24789799 DOI: 10.1128/cvi.00116-14] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Antibodies for the treatment of Clostridium difficile infection (CDI) have been demonstrated to be effective in the research and clinical environments. Early uncertainties about molecular and treatment modalities now appear to have converged upon the systemic dosing of mixtures of human IgG1. Although multiple examples of high-potency monoclonal antibodies (MAbs) exist, significant difficulties were initially encountered in their discovery. This minireview describes historical and contemporary MAbs and highlights differences between the most potent MAbs, which may offer insight into the pathogenesis and treatment of CDI.
Collapse
|
140
|
Recombinant Clostridium difficile toxin fragments as carrier protein for PSII surface polysaccharide preserve their neutralizing activity. Toxins (Basel) 2014; 6:1385-96. [PMID: 24759173 PMCID: PMC4014741 DOI: 10.3390/toxins6041385] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 03/06/2014] [Accepted: 04/09/2014] [Indexed: 11/29/2022] Open
Abstract
Clostridium difficile is a Gram-positive bacterium and is the most commonly diagnosed cause of hospital-associated and antimicrobial-associated diarrhea. Despite the emergence of epidemic C. difficile strains having led to an increase in the incidence of the disease, a vaccine against this pathogen is not currently available. C. difficile strains produce two main toxins (TcdA and TcdB) and express three highly complex cell-surface polysaccharides (PSI, PSII and PSIII). PSII is the more abundantly expressed by most C. difficile ribotypes offering the opportunity of the development of a carbohydrate-based vaccine. In this paper, we evaluate the efficacy, in naive mice model, of PSII glycoconjugates where recombinant toxins A and B fragments (TcdA_B2 and TcdB_GT respectively) have been used as carriers. Both glycoconjugates elicited IgG titers anti-PSII although only the TcdB_GT conjugate induced a response comparable to that obtained with CRM197. Moreover, TcdA_B2 and TcdB_GT conjugated to PSII retained the ability to elicit IgG with neutralizing activity against the respective toxins. These results are a crucial proof of concept for the development of glycoconjugate vaccines against C. difficile infection (CDI) that combine different C. difficile antigens to potentially prevent bacterial colonization of the gut and neutralize toxin activity.
Collapse
|
141
|
Islam J, Taylor AL, Rao K, Huffnagle G, Young VB, Rajkumar C, Cohen J, Papatheodorou P, Aronoff DM, Llewelyn MJ. The role of the humoral immune response to Clostridium difficile toxins A and B in susceptibility to C. difficile infection: a case-control study. Anaerobe 2014; 27:82-6. [PMID: 24708941 DOI: 10.1016/j.anaerobe.2014.03.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 03/25/2014] [Accepted: 03/27/2014] [Indexed: 12/20/2022]
Abstract
Antibody levels to Clostridium difficile toxin A (TcdA), but not toxin B (TcdB), have been found to determine risk of C. difficile infection (CDI). Historically, TcdA was thought to be the key virulence factor; however the importance of TcdB in disease is now established. We re-evaluated the role of antibodies to TcdA and TcdB in determining patient susceptibility to CDI in two separate patient cohorts. In contrast to earlier studies, we find that CDI patients have lower pre-existing IgA titres to TcdB, but not TcdA, when compared to control patients. Our findings suggest that mucosal immunity to TcdB may be important in the early stages of infection and identifies a possible target for preventing CDI progression.
Collapse
Affiliation(s)
- J Islam
- Pathogen Host Interaction Group, Division of Clinical Medicine, Brighton and Sussex Medical School, Medical School Research Building, University of Sussex, Brighton BN1 9PX, UK
| | - A L Taylor
- Pathogen Host Interaction Group, Division of Clinical Medicine, Brighton and Sussex Medical School, Medical School Research Building, University of Sussex, Brighton BN1 9PX, UK
| | - K Rao
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - G Huffnagle
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, USA
| | - V B Young
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, USA
| | - C Rajkumar
- Department of Geriatric Medicine, Royal Sussex County Hospital, Brighton, UK
| | - J Cohen
- Pathogen Host Interaction Group, Division of Clinical Medicine, Brighton and Sussex Medical School, Medical School Research Building, University of Sussex, Brighton BN1 9PX, UK
| | - P Papatheodorou
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albert-Ludwigs-Universität Freiburg, Germany
| | - D M Aronoff
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, USA
| | - M J Llewelyn
- Pathogen Host Interaction Group, Division of Clinical Medicine, Brighton and Sussex Medical School, Medical School Research Building, University of Sussex, Brighton BN1 9PX, UK.
| |
Collapse
|
142
|
Leuzzi R, Adamo R, Scarselli M. Vaccines against Clostridium difficile. Hum Vaccin Immunother 2014; 10:1466-77. [PMID: 24637887 DOI: 10.4161/hv.28428] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Clostridium difficile infection (CDI) is recognized as a major cause of nosocomial diseases ranging from antibiotic related diarrhea to fulminant colitis. Emergence during the last 2 decades of C. difficile strains associated with high incidence, severity and lethal outcomes has increased the challenges for CDI treatment. A limited number of drugs have proven to be effective against CDI and concerns about antibiotic resistance as well as recurring disease solicited the search for novel therapeutic strategies. Active vaccination provides the attractive opportunity to prevent CDI, and intense research in recent years led to development of experimental vaccines, 3 of which are currently under clinical evaluation. This review summarizes recent achievements and remaining challenges in the field of C. difficile vaccines, and discusses future perspectives in view of newly-identified candidate antigens.
Collapse
|
143
|
Spencer J, Leuzzi R, Buckley A, Irvine J, Candlish D, Scarselli M, Douce GR. Vaccination against Clostridium difficile using toxin fragments: Observations and analysis in animal models. Gut Microbes 2014; 5:225-32. [PMID: 24637800 PMCID: PMC4063849 DOI: 10.4161/gmic.27712] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [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
Clostridium difficile is a major cause of antibiotic associated diarrhea. Recently, we have shown that effective protection can be mediated in hamsters through the inclusion of specific recombinant fragments from toxin A and B in a systemically delivered vaccine. Interestingly while neutralizing antibodies to the binding domains of both toxin A and B are moderately protective, enhanced survival is observed when fragments from the glucosyltransferase region of toxin B replace those from the binding domain of this toxin. In this addendum, we discuss additional information that has been derived from such vaccination studies. This includes observations on efficacy and cross-protection against different ribotypes mediated by these vaccines and the challenges that remain for a vaccine which prevents clinical symptoms but not colonization. The use and value of vaccination both in the prevention of infection and for treatment of disease relapse will be discussed.
Collapse
Affiliation(s)
- Janice Spencer
- Institute of Infection, Immunity, and Inflammation; College of Medicine, Veterinary and Life Sciences; University of Glasgow; Glasgow, UK
| | | | - Anthony Buckley
- Institute of Infection, Immunity, and Inflammation; College of Medicine, Veterinary and Life Sciences; University of Glasgow; Glasgow, UK
| | - June Irvine
- Institute of Infection, Immunity, and Inflammation; College of Medicine, Veterinary and Life Sciences; University of Glasgow; Glasgow, UK
| | - Denise Candlish
- Institute of Infection, Immunity, and Inflammation; College of Medicine, Veterinary and Life Sciences; University of Glasgow; Glasgow, UK
| | | | - Gillian R Douce
- Institute of Infection, Immunity, and Inflammation; College of Medicine, Veterinary and Life Sciences; University of Glasgow; Glasgow, UK,Correspondence to: Gillian R Douce,
| |
Collapse
|
144
|
Khanna S, Pardi DS. Clostridium difficile infection: management strategies for a difficult disease. Therap Adv Gastroenterol 2014; 7:72-86. [PMID: 24587820 PMCID: PMC3903088 DOI: 10.1177/1756283x13508519] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Clostridium difficile was first described as a cause of diarrhea in 1978 and in the last three decades has reached an epidemic state with increasing incidence and severity in both healthcare and community settings. There also has been a rise in severe outcomes from C. difficile infection (CDI). There have been tremendous advancements in the field of CDI with the identification of newer risk factors, recognition of CDI in populations previously thought not at risk and development of better diagnostic modalities. Several treatment options are available for CDI apart from metronidazole and vancomycin, and include new drugs such as fidaxomicin and other options such as fecal microbiota transplantation. This review discusses the epidemiology, risk factors and outcomes from CDI, and focuses primarily on existing and evolving treatment modalities.
Collapse
Affiliation(s)
- Sahil Khanna
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| | - Darrell S Pardi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
145
|
Protection from Clostridium difficile infection in CD4 T Cell- and polymeric immunoglobulin receptor-deficient mice. Infect Immun 2013; 82:522-31. [PMID: 24478068 DOI: 10.1128/iai.01273-13] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Clostridium difficile rivals methicillin-resistant Staphylococcus aureus as the primary hospital-acquired infection. C. difficile infection (CDI) caused by toxins A and/or B can manifest as mild diarrhea to life-threatening pseudomembranous colitis. Although most patients recover fully from CDI, ~20% undergo recurrent disease. Several studies have demonstrated a correlation between anti-toxin antibody (Ab) and decreased recurrence; however, the contributions of the systemic and mucosal Ab responses remain unclear. Our goal was to use the CDI mouse model to characterize the protective immune response to C. difficile. C57BL/6 mice infected with epidemic C. difficile strain BI17 developed protective immunity against CDI and did not develop CDI upon rechallenge; they generated systemic IgG and IgA as well as mucosal IgA Ab to toxin. To determine if protective immunity to C. difficile could be generated in immunodeficient individuals, we infected CD4(-/-) mice and found that they generated both mucosal and serum IgA anti-toxin Abs and were protected from CDI upon rechallenge, with protection dependent on major histocompatibility complex class II (MHCII) expression; no IgG anti-toxin Ab was found. We found that protection was likely due to neutralizing mucosal IgA Ab. In contrast, pIgR(-/-) mice, which lack the receptor to transcytose polymeric Ab across the epithelium, were also protected from CDI, suggesting that although mucosal anti-toxin Ab may contribute to protection, it is not required. We conclude that protection from CDI can occur by several mechanisms and that the mechanism of protection is determined by the state of immunocompetence of the host.
Collapse
|
146
|
Yan W, Shin KS, Wang SJ, Xiang H, Divers T, McDonough S, Bowman J, Rowlands A, Akey B, Mohamed H, Chang YF. Equine hyperimmune serum protects mice against Clostridium difficile spore challenge. J Vet Sci 2013; 15:249-58. [PMID: 24136208 PMCID: PMC4087227 DOI: 10.4142/jvs.2014.15.2.249] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 08/14/2013] [Indexed: 01/05/2023] Open
Abstract
Clostridium (C.) difficile is a common cause of nosocomial diarrhea in horses. Vancomycin and metronidazole have been used as standard treatments but are only moderately effective, which highlights the need for a novel alternative therapy. In the current study, we prepared antiserum of equine origin against both C. difficile toxins A and B as well as whole-cell bacteria. The toxin-neutralizing activities of the antibodies were evaluated in vitro and the prophylactic effects of in vivo passive immunotherapy were demonstrated using a conventional mouse model. The data demonstrated that immunized horses generated antibodies against both toxins A and B that possessed toxin-neutralizing activity. Additionally, mice treated with the antiserum lost less weight without any sign of illness and regained weight back to a normal range more rapidly compared to the control group when challenged orally with 107C. difficile spores 1 day after serum injection. These results indicate that intravenous delivery of hyperimmune serum can protect animals from C. difficile challenge in a dose-dependent manner. Hence, immunotherapy may be a promising prophylactic strategy for preventing C. difficile infection in horses.
Collapse
Affiliation(s)
- Weiwei Yan
- Animal Health Diagnostic Center, Departments of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Monaghan TM, Robins A, Knox A, Sewell HF, Mahida YR. Circulating antibody and memory B-Cell responses to C. difficile toxins A and B in patients with C. difficile-associated diarrhoea, inflammatory bowel disease and cystic fibrosis. PLoS One 2013; 8:e74452. [PMID: 24058568 PMCID: PMC3769242 DOI: 10.1371/journal.pone.0074452] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 08/01/2013] [Indexed: 12/17/2022] Open
Abstract
C. difficile infection (CDI) is rarely reported in cystic fibrosis (CF) patients despite frequent hospitalisations and antibiotic usage. Conversely, the prevalence of CDI in inflammatory bowel disease (IBD) has received increased attention. We investigated components of the IgG-specific humoral immune response to C. difficile toxins A and B in patients with C. difficile-associated diarrhoea (CDAD), IBD patients with CDI, CF patients and healthy controls. Serum anti-toxin IgG was determined by ELISA. Circulating antigen-activated B-cells were investigated using Alexa Fluor 488-labelled toxin A and assessed by flow cytometry. Following induction of differentiation of memory B-cells, toxin A- and B-specific antibody secreting cells (ASCs) were quantified using ELISpot. We present the first data showing levels of serum anti-toxin A and B antibodies were significantly higher in patients with CF (without a history of CDI) than in CDAD patients and were stably maintained over time. Notably, the CDAD patients were significantly older than the CF patients. We also show that circulating toxin A-specific memory B-cells (IgD-negative) can be detected in CDAD patients [0.92 (0.09–1.78)%], and were prominent (5.64%, 1.14%) in two CF patients who were asymptomatic carriers of C. difficile. There was correlation between toxin A- and B-specific ASCs, with significantly higher proportions of the latter seen. In some with CDAD, high serum antibody levels were seen to only one of the two toxins. Mucosal secretion of toxin-specific IgG was detected in an additional group of IBD patients with no history of CDI. We conclude that enhanced and stable humoral immune responses to toxins A and B may protect CF and some IBD patients against CDI. The impaired ability to generate strong and/or sustained toxin-specific antibody and memory B-cell responses may increase susceptibility of older patients to CDI and highlight the need to investigate the role of immune senescence in future studies.
Collapse
Affiliation(s)
- Tanya M. Monaghan
- Institute of Infection, Immunity and Inflammation, University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Adrian Robins
- Division of Immunology, University of Nottingham, Nottingham, United Kingdom
| | - Alan Knox
- Division of Respiratory Medicine, University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Herbert F. Sewell
- Division of Immunology, University of Nottingham, Nottingham, United Kingdom
| | - Yashwant R. Mahida
- Institute of Infection, Immunity and Inflammation, University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
- * E-mail:
| |
Collapse
|
148
|
Abstract
Clostridium difficile is a major cause of infection worldwide and is associated with increasing morbidity and mortality in vulnerable patient populations. Metronidazole and oral vancomycin are the currently recommended therapies for the treatment of C. difficile infection (CDI) but are associated with unacceptably high rates of disease recurrence. Novel therapies for the treatment of CDI and prevention of recurrent CDI are urgently needed. Important developments in the treatment of CDI are currently underway and include: novel antibacterial agents with narrower antimicrobial spectra of activity, manipulation of the gut microbiota and enhancement of the host antibody immune response.
Collapse
|
149
|
Lo Vecchio A, Della Ventura B, Nicastro E. Clostridium difficile antibodies: a patent evaluation (WO2013028810). Expert Opin Ther Pat 2013; 23:1635-40. [PMID: 23978053 DOI: 10.1517/13543776.2013.832203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Incidence and severity of Clostridium difficile infection (CDI) are increasing worldwide. Toxins A (TcdA) and B (TcdB) and host immune response are the major determinates of CD pathogenesis and represent a new, stimulating therapeutic target to control CDI. AREAS COVERED The present patent and literature on the pathogenesis and treatment of CD were critically reviewed. The patent was described and put into clinical context, highlighting possible advantages and barriers to use. It consists of a blend of monoclonal antibodies (mAbs) and antigen-binding portions that neutralize TcdA, targeting the enterocyte-binding domain. It demonstrated good efficacy in in vivo models and seems promising in clinical practice. However, recent evidence reshaped the central role of TcdA. EXPERT OPINION Current treatments are inadequate to control CDI and recurrence. Toxin-targeted mAbs are one of the most promising approaches for CDI, including infection by hypervirulent strains. At-risk subjects and those experiencing recurrence are the ideal targets for this second-line treatment; however CDI epidemiology is fast-changing and mAbs may represent a powerful option also for other patients. The re-evaluation of the pathogenic role of TcdA may potentially limit the use of this product; however, the possible administration in combination with other therapeutic agents may optimize its efficacy.
Collapse
Affiliation(s)
- Andrea Lo Vecchio
- University of Naples Federico II, Department of Translational Medical Science, Section of Pediatrics , Via Pansini 5, 80131 Naples , Italy +39 081 7464232 ; +39 081 7464232 ;
| | | | | |
Collapse
|
150
|
Gens KD, Elshaboury RH, Holt JS. Fecal microbiota transplantation and emerging treatments for Clostridium difficile infection. J Pharm Pract 2013; 26:498-505. [PMID: 23966282 DOI: 10.1177/0897190013499527] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Due to the increased incidence and recurrence of Clostridium difficile infection, health care providers are seeking new and alternative treatments to the standard antibiotic therapy. The objective of this article is to present a review on the background, microbiologic efficacy, clinical efficacy, and safety of fecal microbiota transplantation and to provide an overview of emerging treatment options currently under investigation. Emerging treatment options discussed include the use of monoclonal antibodies directed against toxins A and B, C difficile vaccination, and transplantation of nontoxigenic C difficile strains.
Collapse
Affiliation(s)
- Krista D Gens
- Department of Pharmacy, North Memorial Medical Center, Robbinsdale, MN, USA
| | | | | |
Collapse
|