451
|
Status of vaccine research and development for Clostridium difficile. Vaccine 2019; 37:7300-7306. [PMID: 30902484 DOI: 10.1016/j.vaccine.2019.02.052] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/21/2019] [Indexed: 12/15/2022]
Abstract
Clostridium difficile associated disease is fundamentally associated with dysbiosis of the gut microbiome as a consequence of antibiotic use. This is because this sporulating, obligate anaerobe germinates and proliferates rapidly in the dysbiotic gut, which is an indirect consequence of their use. During its growth, C. difficile produces two toxins, toxin A (TcdA) and toxin B (TcdB), which are responsible for the majority of clinical symptoms associated with the disease. Three parenterally delivered vaccines, based on detoxified or recombinant forms of these toxins, have undergone or are undergoing clinical trials. Each offers the opportunity to generate high titres of toxin neutralising antibodies. Whilst these data suggest these vaccines may reduce primary symptomatic disease, they do not in their current form reduce the capacity of the organism to persist and shed from the vaccinated host. The current progress of vaccine development is considered with advantages and limitations of each highlighted. In addition, several alternative approaches are described that seek to limit C. difficile germination, colonisation and persistence. It may yet prove that the most effective treatments to limit infection, disease and spread of the organism will require a combination of therapeutic approaches. The potential use and efficacy of these vaccines in low and middle income countries will be depend on the development of a cost effective vaccine and greater understanding of the distribution and extent of disease in these countries.
Collapse
|
452
|
Mamareli P, Kruse F, Friedrich C, Smit N, Strowig T, Sparwasser T, Lochner M. Epithelium-specific MyD88 signaling, but not DCs or macrophages, control acute intestinal infection with Clostridium difficile. Eur J Immunol 2019; 49:747-757. [PMID: 30802297 DOI: 10.1002/eji.201848022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/21/2019] [Accepted: 02/20/2019] [Indexed: 12/11/2022]
Abstract
Infection with Clostridium difficile is one of the major causes of health care acquired diarrhea and colitis. Signaling though MyD88 downstream of TLRs is critical for initiating the early protective host response in mouse models of C. difficile infection (CDI). In the intestine, MyD88 is expressed in various tissues and cell types, such as the intestinal epithelium and mononuclear phagocytes (MNP), including DC or macrophages. Using a genetic gain-of-function system, we demonstrate here that restricting functional MyD88 signaling to the intestinal epithelium, but also to MNPs is sufficient to protect mice during acute CDI by upregulation of the intestinal barrier function and recruitment of neutrophils. Nevertheless, we also show that mice depleted for CD11c-expressing MNPs in the intestine display no major defects in mounting an effective inflammatory response, indicating that the absence of these cells is irrelevant for inducing host protection during acute infection. Together, our results highlight the importance of epithelial-specific MyD88 signaling and demonstrate that although functional MyD88 signaling in DC and macrophages alone is sufficient to correct the phenotype of MyD88-deficiency, these cells do not seem to be essential for host protection in MyD88-sufficient animals during acute infection with C. difficile.
Collapse
Affiliation(s)
- Panagiota Mamareli
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany.,Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Friederike Kruse
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Christin Friedrich
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany.,Institute of Medical Microbiology and Hygiene, University of Freiburg, Freiburg, Germany.,Institute of Systems Immunology, University of Würzburg, Würzburg, Germany
| | - Nathiana Smit
- Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Till Strowig
- Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany.,Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Matthias Lochner
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| |
Collapse
|
453
|
Aziz M, Fatima R, Douglass LN, Abughanimeh O, Raza S. Current updates in management of Clostridium difficile infection in cancer patients. Curr Med Res Opin 2019; 35:473-478. [PMID: 29888965 DOI: 10.1080/03007995.2018.1487389] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Clostridium difficile infection (CDI) is a significant health burden, now recognized as the leading cause of acquired diarrhea in patients receiving antibiotic therapy. Complications of infection with this pathogen include severe diarrhea, causing electrolyte imbalances, dehydration, hemodynamic instability, toxic megacolon, shock, and death. Hence it is extremely paramount to stay updated on management options for this infection, especially in cancer patients. REVIEW This article presents an in-depth review of literature on the treatment modalities available for CDI in cancer patients. Relevant articles highlighting therapeutic and symptomatic management of CDI patients with underlying malignancy have been summarized. CONCLUSIONS Despite the current options available, more studies are needed to assess the newer therapeutic options that are being employed for populations other than cancer patients.
Collapse
Affiliation(s)
- Muhammad Aziz
- a Department of Internal Medicine , University of Kansas Medical Center , Kansas City , MO , USA
| | - Rawish Fatima
- b Department of Medicine , Dow University of Health Sciences , Karachi , Pakistan
| | - Lindsey N Douglass
- c Pharmacy Department , Saint Luke's Hospital of Kansas City , Kansas City , MO , USA
| | - Omar Abughanimeh
- d Department of Internal Medicine , Saint Luke's Hospital of Kansas City/University of Missouri , Kansas City , MO , USA
| | - Shahzad Raza
- e Department of Hematology & Oncology , St. Luke's Hospital of Kansas City/University of Missouri , Kansas City , MO , USA
| |
Collapse
|
454
|
Mcilroy JR, Segal JP, Mullish BH, Nabil Quraishi M, Gasbarrini A, Cammarota G, Ianiro G. Current and future targets for faecal microbiota transplantation. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.humic.2018.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
455
|
Belete TM. Novel targets to develop new antibacterial agents and novel alternatives to antibacterial agents. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.humic.2019.01.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
456
|
Sartelli M, Di Bella S, McFarland LV, Khanna S, Furuya-Kanamori L, Abuzeid N, Abu-Zidan FM, Ansaloni L, Augustin G, Bala M, Ben-Ishay O, Biffl WL, Brecher SM, Camacho-Ortiz A, Caínzos MA, Chan S, Cherry-Bukowiec JR, Clanton J, Coccolini F, Cocuz ME, Coimbra R, Cortese F, Cui Y, Czepiel J, Demetrashvili Z, Di Carlo I, Di Saverio S, Dumitru IM, Eckmann C, Eiland EH, Forrester JD, Fraga GP, Frossard JL, Fry DE, Galeiras R, Ghnnam W, Gomes CA, Griffiths EA, Guirao X, Ahmed MH, Herzog T, Kim JI, Iqbal T, Isik A, Itani KMF, Labricciosa FM, Lee YY, Juang P, Karamarkovic A, Kim PK, Kluger Y, Leppaniemi A, Lohsiriwat V, Machain GM, Marwah S, Mazuski JE, Metan G, Moore EE, Moore FA, Ordoñez CA, Pagani L, Petrosillo N, Portela F, Rasa K, Rems M, Sakakushev BE, Segovia-Lohse H, Sganga G, Shelat VG, Spigaglia P, Tattevin P, Tranà C, Urbánek L, Ulrych J, Viale P, Baiocchi GL, Catena F. 2019 update of the WSES guidelines for management of Clostridioides ( Clostridium) difficile infection in surgical patients. World J Emerg Surg 2019; 14:8. [PMID: 30858872 PMCID: PMC6394026 DOI: 10.1186/s13017-019-0228-3] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/17/2019] [Indexed: 02/08/2023] Open
Abstract
In the last three decades, Clostridium difficile infection (CDI) has increased in incidence and severity in many countries worldwide. The increase in CDI incidence has been particularly apparent among surgical patients. Therefore, prevention of CDI and optimization of management in the surgical patient are paramount. An international multidisciplinary panel of experts from the World Society of Emergency Surgery (WSES) updated its guidelines for management of CDI in surgical patients according to the most recent available literature. The update includes recent changes introduced in the management of this infection.
Collapse
Affiliation(s)
- Massimo Sartelli
- Department of Surgery, Macerata Hospital, Via Santa Lucia 2, 62100 Macerata, Italy
| | - Stefano Di Bella
- Infectious Diseases Department, Trieste University Hospital, Trieste, Italy
| | - Lynne V. McFarland
- Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA USA
| | - Sahil Khanna
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN USA
| | - Luis Furuya-Kanamori
- Research School of Population Health, Australian National University, Acton, ACT Australia
| | - Nadir Abuzeid
- Department of Microbiology, Faculty of Medical Laboratory Sciences, Omdurman Islamic University, Khartoum, Sudan
| | - Fikri M. Abu-Zidan
- Department of Surgery, College of Medicine and Health Sciences, UAE University, Al-Ain, United Arab Emirates
| | - Luca Ansaloni
- Department of General Surgery, Bufalini Hospital, Cesena, Italy
| | - Goran Augustin
- Department of Surgery, University Hospital Centre Zagreb and School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Miklosh Bala
- Trauma and Acute Care Surgery Unit, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Offir Ben-Ishay
- Department of General Surgery, Rambam Health Care Campus, Haifa, Israel
| | - Walter L. Biffl
- Trauma and Acute Care Surgery, Scripps Memorial Hospital La Jolla, La Jolla, CA USA
| | - Stephen M. Brecher
- Pathology and Laboratory Medicine, VA Boston Healthcare System, West Roxbury MA and BU School of Medicine, Boston, MA USA
| | - Adrián Camacho-Ortiz
- Department of Internal Medicine, University Hospital, Dr. José E. González, Monterrey, Mexico
| | - Miguel A. Caínzos
- Department of Surgery, University of Santiago de Compostela, A Coruña, Spain
| | - Shirley Chan
- Department of General Surgery, Medway Maritime Hospital, Gillingham, Kent UK
| | - Jill R. Cherry-Bukowiec
- Department of Surgery, Division of Acute Care Surgery, University of Michigan, Ann Arbor, MI USA
| | - Jesse Clanton
- Department of Surgery, West Virginia University Charleston Division, Charleston, WV USA
| | | | - Maria E. Cocuz
- Faculty of Medicine, Transilvania University, Infectious Diseases Hospital, Brasov, Romania
| | - Raul Coimbra
- Riverside University Health System Medical Center and Loma Linda University School of Medicine, Moreno Valley, CA USA
| | | | - Yunfeng Cui
- Department of Surgery, Tianjin Nankai Hospital, Nankai Clinical School of Medicine, Tianjin Medical University, Tianjin, China
| | - Jacek Czepiel
- Department of Infectious Diseases, Jagiellonian University, Medical College, Kraków, Poland
| | - Zaza Demetrashvili
- Department of Surgery, Tbilisi State Medical University, Kipshidze Central University Hospital, Tbilisi, Georgia
| | - Isidoro Di Carlo
- Department of Surgical Sciences, Cannizzaro Hospital, University of Catania, Catania, Italy
| | - Salomone Di Saverio
- Department of Surgery, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Irina M. Dumitru
- Clinical Infectious Diseases Hospital, Ovidius University, Constanta, Romania
| | - Christian Eckmann
- Department of General, Visceral and Thoracic Surgery, Klinikum Peine, Hospital of Medical University Hannover, Peine, Germany
| | | | | | - Gustavo P. Fraga
- Division of Trauma Surgery, Hospital de Clinicas, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Jean L. Frossard
- Service of Gastroenterology and Hepatology, Geneva University Hospital, Genève, Switzerland
| | - Donald E. Fry
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL USA
- University of New Mexico School of Medicine, Albuquerque, NM USA
| | - Rita Galeiras
- Critical Care Unit, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), A Coruña, Spain
| | - Wagih Ghnnam
- Department of Surgery Mansoura, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Carlos A. Gomes
- Surgery Department, Hospital Universitario (HU) Terezinha de Jesus da Faculdade de Ciencias Medicas e da Saude de Juiz de Fora (SUPREMA), Hospital Universitario (HU) Universidade Federal de Juiz de Fora (UFJF), Juiz de Fora, Brazil
| | | | - Xavier Guirao
- Unit of Endocrine, Head, and Neck Surgery and Unit of Surgical Infections Support, Department of General Surgery, Parc Taulí, Hospital Universitari, Sabadell, Spain
| | - Mohamed H. Ahmed
- Department of Medicine, Milton Keynes University Hospital NHS Foundation Trust, Milton Keynes, Buckinghamshire UK
| | - Torsten Herzog
- Department of Surgery, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Jae Il Kim
- Department of Surgery, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Republic of Korea
| | - Tariq Iqbal
- Department of Gastroenterology, Queen Elizabeth Hospital, Birmingham, UK
| | - Arda Isik
- General Surgery Department, Magee Womens Hospital, UPMC, Pittsburgh, USA
| | - Kamal M. F. Itani
- Department of Surgery, VA Boston Health Care System, Boston University and Harvard Medical School, Boston, MA USA
| | | | - Yeong Y. Lee
- School of Medical Sciences, University Sains Malaysia, Kota Bharu, Kelantan Malaysia
| | - Paul Juang
- Department of Pharmacy Practice, St Louis College of Pharmacy, St Louis, MO USA
| | - Aleksandar Karamarkovic
- Faculty of Mediine University of Belgrade Clinic for Surgery “Nikola Spasic”, University Clinical Center “Zvezdara” Belgrade, Belgrade, Serbia
| | - Peter K. Kim
- Department of Surgery, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY USA
| | - Yoram Kluger
- Department of General Surgery, Rambam Health Care Campus, Haifa, Israel
| | - Ari Leppaniemi
- Abdominal Center, Helsinki University Hospital Meilahti, Helsinki, Finland
| | - Varut Lohsiriwat
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Gustavo M. Machain
- Department of Surgery, Universidad Nacional de Asuncion, Asuncion, Paraguay
| | - Sanjay Marwah
- Department of Surgery, Post-Graduate Institute of Medical Sciences, Rohtak, India
| | - John E. Mazuski
- Department of Surgery, Washington University School of Medicine, Saint Louis, USA
| | - Gokhan Metan
- Department of Infectious Diseases and Clinical Microbiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Ernest E. Moore
- Department of Surgery, University of Colorado, Denver Health Medical Center, Denver, CO USA
| | | | - Carlos A. Ordoñez
- Department of Surgery, Fundación Valle del Lili, Hospital Universitario del Valle, Universidad del Valle, Cali, Colombia
| | - Leonardo Pagani
- Infectious Diseases Unit, Bolzano Central Hospital, Bolzano, Italy
| | - Nicola Petrosillo
- National Institute for Infectious Diseases - INMI - Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Francisco Portela
- Gastroenterology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Kemal Rasa
- Department of Surgery, Anadolu Medical Center, Kocaali, Turkey
| | - Miran Rems
- Department of Abdominal and General Surgery, General Hospital Jesenice, Jesenice, Slovenia
| | | | | | - Gabriele Sganga
- Division of Emergency Surgery, Department of Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Vishal G. Shelat
- Department of Surgery, Tan Tock Seng Hospital, Singapore, Singapore
| | - Patrizia Spigaglia
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Pierre Tattevin
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, Rennes, France
| | - Cristian Tranà
- Department of Surgery, Macerata Hospital, Via Santa Lucia 2, 62100 Macerata, Italy
| | - Libor Urbánek
- First Department of Surgery, Faculty of Medicine, Masaryk University Brno and University Hospital of St. Ann Brno, Brno, Czech Republic
| | - Jan Ulrych
- First Department of Surgery, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - Pierluigi Viale
- Clinic of Infectious Diseases, St Orsola-Malpighi University Hospital, Bologna, Italy
| | - Gian L. Baiocchi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Fausto Catena
- Emergency Surgery Department, Maggiore Parma Hospital, Parma, Italy
| |
Collapse
|
457
|
Tran MCN, Kullar R, Goldstein EJC. Investigational drug therapies currently in early-stage clinical development for the treatment of clostridioides (clostridium) difficile infection. Expert Opin Investig Drugs 2019; 28:323-335. [DOI: 10.1080/13543784.2019.1581763] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Mai-Chi N. Tran
- Department of Pharmacy, Providence St. John’s Health Center, Santa Monica,
CA, USA
- Department of Pharmacy, Clinica Juan Pablo Medical Group, Los Angeles,
CA, USA
| | | | - Ellie J. C. Goldstein
- R M Alden Research Laboratory, Santa Monica,
CA, USA
- David Geffen School of Medicine, Los Angeles,
CA, USA
| |
Collapse
|
458
|
|
459
|
Leslie JL, Vendrov KC, Jenior ML, Young VB. The Gut Microbiota Is Associated with Clearance of Clostridium difficile Infection Independent of Adaptive Immunity. mSphere 2019; 4:e00698-18. [PMID: 30700514 PMCID: PMC6354811 DOI: 10.1128/mspheredirect.00698-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 01/05/2019] [Indexed: 12/26/2022] Open
Abstract
Clostridium (Clostridioides) difficile, a Gram-positive, anaerobic bacterium, is the leading single cause of nosocomial infections in the United States. A major risk factor for Clostridium difficile infection (CDI) is prior exposure to antibiotics, as they increase susceptibility to CDI by altering the membership of the microbial community enabling colonization. The importance of the gut microbiota in providing protection from CDI is underscored by the reported 80 to 90% success rate of fecal microbial transplants in treating recurrent infections. Adaptive immunity, specifically humoral immunity, is also sufficient to protect from both acute and recurrent CDI. However, the role of the adaptive immune system in mediating clearance of C. difficile has yet to be resolved. Using murine models of CDI, we found that adaptive immunity is dispensable for clearance of C. difficile However, random forest analysis using only two members of the resident bacterial community correctly identified animals that would go on to clear the infection with 66.7% accuracy. These findings indicate that the indigenous gut microbiota independent of adaptive immunity facilitates clearance of C. difficile from the murine gastrointestinal tract.IMPORTANCEClostridium difficile infection is a major cause of morbidity and mortality in hospitalized patients in the United States. Currently, the role of the adaptive immune response in modulating levels of C. difficile colonization is unresolved. This work suggests that the indigenous gut microbiota is a main factor that promotes clearance of C. difficile from the GI tract. Our results show that clearance of C. difficile can occur without contributions from the adaptive immune response. This study also has implications for the design of preclinical studies testing the efficacy of vaccines on clearance of bacterial pathogens, as inherent differences in the baseline community structure of animals may bias findings.
Collapse
Affiliation(s)
- Jhansi L Leslie
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Kimberly C Vendrov
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Matthew L Jenior
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Vincent B Young
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
460
|
Guery B. Clostridium difficile infection trials: what is the primary endpoint? THE LANCET. INFECTIOUS DISEASES 2019; 19:219-220. [PMID: 30709664 DOI: 10.1016/s1473-3099(18)30626-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 10/10/2018] [Indexed: 11/25/2022]
Affiliation(s)
- Benoit Guery
- Infectious Diseases Service, Department of Medicine, University Hospital and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
461
|
Gerding DN, Cornely OA, Grill S, Kracker H, Marrast AC, Nord CE, Talbot GH, Buitrago M, Gheorghe Diaconescu I, Murta de Oliveira C, Preotescu L, Pullman J, Louie TJ, Wilcox MH. Cadazolid for the treatment of Clostridium difficile infection: results of two double-blind, placebo-controlled, non-inferiority, randomised phase 3 trials. THE LANCET. INFECTIOUS DISEASES 2019; 19:265-274. [PMID: 30709665 DOI: 10.1016/s1473-3099(18)30614-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/06/2018] [Accepted: 10/02/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cadazolid is a novel quinoxolidinone antibiotic developed for treating Clostridium difficile infection. We aimed to investigate the safety and efficacy of cadazolid compared with vancomycin in patients with C difficile infection. METHODS IMPACT 1 and IMPACT 2 were identically designed, multicentre, double-blind, placebo-controlled, non-inferiority, randomised phase 3 trials. IMPACT 1 was done in Australia, Brazil, Canada, France, Germany, Italy, the Netherlands, Peru, Poland, Romania, Spain, and the USA, and IMPACT 2 was done in Argentina, Belgium, Brazil, Canada, Chile, Croatia, Czech Republic, Greece, Hungary, Israel, Romania, Slovakia, South Korea, the UK, and the USA. Patients (aged 18 years or older) with mild-to-moderate or severe C difficile infection (diarrhoea with positive glutamate dehydrogenase and toxin A or B enzyme immunoassays) were randomly assigned (1:1) with a randomisation list stratified by centre and C difficile infection episode type (block size of four), and allocation was masked to investigators and participants. Patients received either oral cadazolid 250 mg twice daily with vancomycin-matching placebo capsule four times daily or oral vancomycin 125 mg four times a day with cadazolid-matching placebo suspension twice daily for 10 days, with 30 days of follow-up. The primary efficacy outcome was non-inferiority (margin -10%) of cadazolid versus vancomycin for clinical cure in the modified intention-to-treat and per-protocol populations. Clinical cure was defined as resolution of diarrhoea with no additional treatment for C difficile infection. These trials are registered with ClinicalTrials.gov, numbers NCT01987895 (IMPACT 1) and NCT01983683 (IMPACT 2). FINDINGS Between March 28, 2014, and March 24, 2017, for IMPACT 1, and Dec 13, 2013, and May 2, 2017, for IMPACT 2, 1263 participants were randomly assigned to receive cadazolid (306 in IMPACT 1 and 298 in IMPACT 2) or vancomycin (326 in IMPACT 1 and 311 in IMPACT 2). In the modified intention-to-treat population in IMPACT 1, 253 (84%) of 302 had clinical cure in the cadazolid group versus 271 (85%) of 318 in the vancomycin group. In IMPACT 2, 235 (81%) of 290 versus 258 (86%) of 301 had clinical cure. In the per-protocol population, 247 (88%) of 282 versus 264 (92%) of 288 had clinical cure in IMPACT 1 and 214 (87%) of 247 versus 237 (92%) of 259 in IMPACT 2. Non-inferiority for clinical cure to vancomycin was shown in IMPACT 1 but not in IMPACT 2 (IMPACT 1 treatment difference: -1·4 [95% CI -7·2 to 4·3] for modified intention to treat and -4·1 [-9·2 to 1·0] for per protocol; IMPACT 2: -4·7 [-10·7 to 1·3] for modified intention to treat and -4·9 [-10·4 to 0·6] for per protocol). The safety and tolerability profiles of the two antibiotics were similar. INTERPRETATION Cadazolid was safe and well tolerated but did not achieve its primary endpoint of non-inferiority to vancomycin for clinical cure in one of two phase 3 C difficile infection trials. Therefore, further commercial development of cadazolid for C difficile infection is unlikely. FUNDING Actelion Pharmaceuticals.
Collapse
Affiliation(s)
- Dale N Gerding
- Edward Hines Jr Veterans Administration Hospital, Hines, IL, USA.
| | - Oliver A Cornely
- Department of Internal Medicine, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Clinical Trials Centre Cologne, University of Cologne, Cologne, Germany
| | - Simon Grill
- Actelion Pharmaceuticals, Allschwil, Switzerland
| | | | | | - Carl Erik Nord
- Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | | | - Liliana Preotescu
- Matei Bals National Institute of Infectious Diseases, Bucharest, Romania
| | | | - Thomas J Louie
- Foothills Medical Center, Alberta Health Services & University of Calgary, Cumming School of Medicine, Calgary, AB, Canada
| | - Mark H Wilcox
- Microbiology, Old Medical School, Leeds General Infirmary, Leeds, UK
| |
Collapse
|
462
|
Fatima R, Aziz M. The Hypervirulent Strain of Clostridium Difficile: NAP1/B1/027 - A Brief Overview. Cureus 2019; 11:e3977. [PMID: 30967977 PMCID: PMC6440555 DOI: 10.7759/cureus.3977] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 01/29/2019] [Indexed: 12/20/2022] Open
Abstract
Clostridium difficile is a gram-positive bacterium notorious for causing epidemic diarrhea globally with a significant health burden. The pathogen is clinically challenging with increasing antibiotic resistance and recurrence rate. We provide here an in-depth review of one particular strain/ribotype 027, commonly known as NAP1/B1/027 or North American pulsed-field gel electrophoresis type 1, restriction endonuclease analysis type B1, polymerase chain reaction ribotype 027, which has shown a much higher recurrence rate than other strains.
Collapse
Affiliation(s)
- Rawish Fatima
- Internal Medicine, Dow University of Health Sciences, Karachi, PAK
| | - Muhammad Aziz
- Internal Medicine, University of Kansas Medical Center, Kansas City, USA
| |
Collapse
|
463
|
Population Pharmacokinetics and Pharmacodynamics of Bezlotoxumab in Adults with Primary and Recurrent Clostridium difficile Infection. Antimicrob Agents Chemother 2019; 63:AAC.01971-18. [PMID: 30455246 DOI: 10.1128/aac.01971-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/08/2018] [Indexed: 12/18/2022] Open
Abstract
The fully human monoclonal antibody bezlotoxumab is indicated for preventing the recurrence of Clostridioides difficile (formerly Clostridium difficile) infection (CDI) in adults who receive antibacterial treatment for CDI and who are at high risk for a CDI recurrence. The efficacy and safety of 10-mg/kg of body weight bezlotoxumab were demonstrated in two phase 3 trials: the MODIFY I (ClinicalTrials.gov registration number NCT01241552) and MODIFY II (ClinicalTrials.gov registration number NCT01513239) trials. Here, a population pharmacokinetic (popPK) analysis, performed using data from the MODIFY I and II trials (n = 1,515) and from three phase 1 trials (n = 72) to characterize bezlotoxumab pharmacokinetics (PK) in phase 3 clinical trial participants and in healthy subjects, is reported. A stepwise covariate search was conducted to identify factors influencing PK. Post hoc-estimated bezlotoxumab exposures from the popPK model were used to conduct an exposure-response analysis for CDI recurrence. Bezlotoxumab PK were described by a two-compartment model with linear elimination and allometric scaling for clearance and the volume of distribution by body weight. Although the final popPK model included gender, ethnicity (Japanese descent), race (black versus nonblack), and albumin level as significant covariates, the impact of these factors was not clinically meaningful, based on the totality of the PK and clinical experience. Exposure-response analysis of CDI recurrence demonstrated a similar low rate of CDI recurrence over the entire range of exposures achieved in the phase 3 trials, indicating that exposures were on the maximal response plateau of the exposure-response curve. Overall, the analyses confirmed the appropriateness of the 10-mg/kg dose across the phase 3 trial population with no dose adjustments necessary over a broad demographic background.
Collapse
|
464
|
Icho S, Melnyk RA. Dismantling a Toxin to Disarm a Superbug. Trends Pharmacol Sci 2019; 40:155-156. [PMID: 30685063 DOI: 10.1016/j.tips.2019.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 01/14/2019] [Indexed: 10/27/2022]
Abstract
Clostridium difficile (Clostridioides difficile) is a toxin-producing, multidrug-resistant bacterium. Inhibiting the effects of toxins, which are responsible for the symptoms of disease, is viewed as a promising non-antibiotic approach to treat C. difficile infection (CDI). By inducing premature activation of toxins, Ivarsson and colleagues (Cell Chemical Biology 2018; http://doi.org/10.1016/j.chembiol.2018.10.002) uncover a clever new strategy to block toxin action.
Collapse
Affiliation(s)
- Simoun Icho
- Molecular Medicine, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Roman A Melnyk
- Molecular Medicine, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
465
|
Abstract
Bezlotoxumab (Zinplava™) is a fully human monoclonal antibody against Clostridium difficile toxin B indicated for the prevention of C. difficile infection (CDI) recurrence in patients with a high recurrence risk. It is the first agent approved for recurrence prevention and is administered as a single intravenous infusion in conjunction with standard-of-care (SoC) antibacterial treatment for CDI. In well-designed, placebo-controlled, phase 3 trials (MODIFY 1 and 2), a single infusion of bezlotoxumab, given in combination with SoC antibacterial therapy for CDI in adults, was effective in reducing CDI recurrence in the 12 weeks post-treatment, with this benefit being seen mainly in the patients at high recurrence risk. Bezlotoxumab did not impact the efficacy of the antibacterials being used to treat the CDI and, consistent with its benefits on CDI recurrence, appeared to reduce the need for subsequent antibacterials, thus minimizing further gut microbiota disruption. Longer term, there were no further CDI recurrences over 12 months' follow-up among patients who had received bezlotoxumab in MODIFY 2 and entered an extension substudy. Bezlotoxumab has low immunogenicity and is generally well tolerated, although the potential for heart failure in some patients requires consideration; cost-effectiveness data for bezlotoxumab are awaited with interest. Thus, a single intravenous infusion of bezlotoxumab during SoC antibacterial treatment for CDI is an emerging option for reducing CDI recurrence in adults at high risk of recurrence.
Collapse
|
466
|
Hypoalbuminemia as predictor of recurrence of Clostridium difficile infection. Wien Klin Wochenschr 2019; 131:68-74. [PMID: 30617709 PMCID: PMC6394683 DOI: 10.1007/s00508-018-1432-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/06/2018] [Indexed: 02/07/2023]
Abstract
Background Novel drugs for Clostridium difficile (C. difficile) infections have been proven to reduce recurrent infections. Because of their high financial costs, identification of patients at high risk for recurrence is essential to provide optimal treatment. The ATLAS score’s ability to predict 90-day recurrence, disease complications and 1‑year all-cause mortality was evaluated. Methods 144 consecutive symptomatic patients with positive stool test for C. difficile were enrolled. The ATLAS score (consisting of the variables age, temperature, leukocyte count, albumin, systemic antibiotics, serum creatinine) was calculated and patients were stratified into 4 subgroups according to their scores. A Cox regression model was used to estimate the extent to which ATLAS was associated with 90-day recurrence. Furthermore, the score was correlated with disease complications and one-year all-cause mortality. Results ATLAS was unable to predict 90-day recurrence (p = 0.064, HR 1.134 [0.993;1.295]), but performed well for disease complications (D = 0.382, p < 0.001, HR 1.547 [1.266;1.889]) and mortality (p < 0.001, HR 1.374 [1.194;1.583]). Serum albumin was the only parameter able to predict 90-day recurrence (p = 0.016, HR 0.958 [0.926;0.992]) and was also a predictor of disease complications (p < 0.001, HR 0.865[0.809;0.924]) and one-year all-cause mortality (p < 0.001, HR 0.923 [0.896;0.950]). A threshold of 33.1g/L (sensitivity = 56%, specificity = 80%, AUC 0.683) and 29.2g/L (sensitivity = 75%, specificity = 70%, AUC 0.763) of serum albumin could be identified to be predictive for 90-day recurrence and one-year all-cause mortality, respectively. Conclusions Serum albumin and ATLAS are predictors of disease complications and mortality, while only serum albumin is significantly associated with 90-day disease recurrence.
Collapse
|
467
|
Orenstein R, Patron RL. Clostridioides difficile therapeutics: guidelines and beyond. Ther Adv Infect Dis 2019; 6:2049936119868548. [PMID: 31448117 PMCID: PMC6693025 DOI: 10.1177/2049936119868548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/17/2019] [Indexed: 12/21/2022] Open
Abstract
Clostridioides difficile infection (CDI) has become an increasingly common infection both within and outside of the hospital setting. The management of this infection has been evolving as we learn more about the role of the human microbiota in protecting us from this gastrointestinal opportunist. For many years the focus of treatment had been on eradication of the vegetative, toxin-producing form of the organism, with little regard for its collateral impact on the host's microbiota or risk of recurrence. With the marked increase in C. difficile disease, and, particularly, recurrent disease in the last decade, new guidelines are more focused on targeting and reducing collateral damage to the colonic microbiota. Immune-based strategies that manipulate the microbiota and provide a humoral response to toxins have now become mainstream. Newer strategies are needed to look beyond simply resolving the primary episode but are focused on delayed outcomes such as cure at 90 days, reduced morbidity and mortality, and patient quality of life. The purpose of this review is to familiarize readers with the most recent evidence-based guidelines for C. difficile management, and to describe the role of newer antimicrobials, immunological-, and microbiota-based therapeutics to prevent recurrence and improve the outcomes of people with CDI.
Collapse
Affiliation(s)
- Robert Orenstein
- Division of Infectious Diseases, Mayo Clinic
Arizona, 5777 East Mayo Boulevard, Phoenix, AZ 85054, USA
| | - Roberto L. Patron
- Division of Infectious Diseases, Mayo Clinic
Arizona, Phoenix, AZ, USA
| |
Collapse
|
468
|
Dieterle MG, Rao K, Young VB. Novel therapies and preventative strategies for primary and recurrent Clostridium difficile infections. Ann N Y Acad Sci 2019; 1435:110-138. [PMID: 30238983 PMCID: PMC6312459 DOI: 10.1111/nyas.13958] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/16/2018] [Accepted: 08/03/2018] [Indexed: 12/15/2022]
Abstract
Clostridium difficile is the leading infectious cause of antibiotic-associated diarrhea and colitis. C. difficile infection (CDI) places a heavy burden on the healthcare system, with nearly half a million infections yearly and an approximate 20% recurrence risk after successful initial therapy. The high incidence has driven new research on improved prevention such as the emerging use of probiotics, intestinal microbiome manipulation during antibiotic therapies, vaccinations, and newer antibiotics that reduce the disruption of the intestinal microbiome. While the treatment of acute C. difficile is effective in most patients, it can be further optimized by adjuvant therapies that improve the initial treatment success and decrease the risk of subsequent recurrence. Finally, the high risk of recurrence has led to multiple emerging therapies that target toxin activity, recovery of the intestinal microbial community, and elimination of latent C. difficile in the intestine. In summary, CDIs illustrate the complex interaction among host physiology, microbial community, and pathogen that requires specific therapies to address each of the factors leading to primary infection and recurrence.
Collapse
Affiliation(s)
- Michael G. Dieterle
- University of Michigan Medical School, Medical Scientist Training Program (MSTP), Ann Arbor, Michigan
- University of Michigan Department of Microbiology and Immunology, Ann Arbor, Michigan
| | - Krishna Rao
- University of Michigan Department of Internal Medicine, Infectious Diseases Division, Ann Arbor, Michigan
| | - Vincent B. Young
- University of Michigan Department of Microbiology and Immunology, Ann Arbor, Michigan
- University of Michigan Department of Internal Medicine, Infectious Diseases Division, Ann Arbor, Michigan
| |
Collapse
|
469
|
Krajicek E, Fischer M, Allegretti JR, Kelly CR. Nuts and Bolts of Fecal Microbiota Transplantation. Clin Gastroenterol Hepatol 2019; 17:345-352. [PMID: 30268564 DOI: 10.1016/j.cgh.2018.09.029] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/14/2018] [Accepted: 09/19/2018] [Indexed: 02/07/2023]
Abstract
Clostridium difficile infection (CDI) has become the leading cause of nosocomial infection in the United States with significant risk of both morbidity and mortality. While antimicrobial therapy forms the basis of treatment, there are several clinical scenarios in which antimicrobial therapy alone is insufficient. Evidence continues to show the safety and efficacy fecal microbiota transplantation (FMT) in recurrent and severe CDI. This review will outline FMT efficacy, safety, and indications and present practical advice for clinicians interested in best practices around delivery of FMT.
Collapse
Affiliation(s)
- Edward Krajicek
- Division of Gastroenterology, Indiana University, Indianapolis, Indiana
| | - Monika Fischer
- Division of Gastroenterology, Indiana University, Indianapolis, Indiana
| | - Jessica R Allegretti
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Colleen R Kelly
- Division of Gastroenterology, Alpert Medical School of Brown University, Providence, Rhode Island.
| |
Collapse
|
470
|
Ivarsson ME, Durantie E, Huberli C, Huwiler S, Hegde C, Friedman J, Altamura F, Lu J, Verdu EF, Bercik P, Logan SM, Chen W, Leroux JC, Castagner B. Small-Molecule Allosteric Triggers of Clostridium difficile Toxin B Auto-proteolysis as a Therapeutic Strategy. Cell Chem Biol 2019; 26:17-26.e13. [DOI: 10.1016/j.chembiol.2018.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 06/27/2018] [Accepted: 09/28/2018] [Indexed: 01/19/2023]
|
471
|
Pelfrene E, Mura M, Cavaleiro Sanches A, Cavaleri M. Monoclonal antibodies as anti-infective products: a promising future? Clin Microbiol Infect 2019; 25:60-64. [PMID: 29715552 PMCID: PMC7128139 DOI: 10.1016/j.cmi.2018.04.024] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/19/2018] [Accepted: 04/23/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND The paucity of licensed monoclonal antibodies (mAbs) in the infectious diseases arena strongly contrasts with the ready availability of these therapeutics for use in other conditions. AIMS This narrative review aims to assess the potential of monoclonal antibody-based interventions for infectious diseases. SOURCES A review of the literature via the Medline database was performed and complemented by published official documents on licensed anti-infective mAbs. In addition, ongoing trials were identified through a search of the clinical trial registration platform ClinicalTrials.gov. CONTENT We identified the few infections for which mAbs have been added to the therapeutic armamentarium and stressed their potential in representing a readily available protection tool against biothreats and newly emerging and reemerging infectious agents. In reviewing the historical context and main features of mAbs, we assert a potentially wider applicability and cite relevant examples of ongoing therapeutic developments. Factors hindering successful introduction of mAbs on a larger scale are outlined and thoughts are offered on how to possibly address some of these limitations. IMPLICATIONS mAbs may represent important tools in treating or preventing infections occurring with reasonably sufficient prevalence to justify demand and for which existing alternatives are not deemed fully adequate. Future initiatives need to address the prohibitive costs encountered in the development process. The feasibility of more large-scale administration of alternative modalities merits further exploration. In order to ensure optimal prospect of regulatory success, an early dialogue with competent authorities is encouraged.
Collapse
Affiliation(s)
- E Pelfrene
- Office of Anti-infectives and Vaccines, Human Medicines Evaluation Division, European Medicines Agency, London, UK.
| | - M Mura
- Office of Anti-infectives and Vaccines, Human Medicines Evaluation Division, European Medicines Agency, London, UK
| | - A Cavaleiro Sanches
- Quality Office, Human Medicines Research & Development Support Division, European Medicines Agency, London, UK
| | - M Cavaleri
- Office of Anti-infectives and Vaccines, Human Medicines Evaluation Division, European Medicines Agency, London, UK
| |
Collapse
|
472
|
Analysis of Treatment Outcomes for Recurrent Clostridium difficile Infections and Fecal Microbiota Transplantation in a Pediatric Hospital. Pediatr Infect Dis J 2019; 38:32-36. [PMID: 29601446 DOI: 10.1097/inf.0000000000002053] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Clostridium difficile infection (CDI) is one of the most common nosocomial infections in the United States, with an increasing incidence in children. Approximately 20% of pediatric patients develop recurrent infections. It's imperative to further analyze the incidence of recurrent CDI in the pediatric population and determine the most effective treatments. The primary goal of this study is to characterize children with recurrent CDI at our institution, including both hospital-acquired CDI (HA-CDI) and community-acquired CDI (CA-CDI) cases, summarize the various treatments utilized, including fecal microbiota transplant (FMT) and compare their success rates. METHODS A retrospective cohort study of pediatric patients 1-21 years of age treated for CDI at a single institution from January 2010 to December 2014 was performed. RESULTS There were 175 subjects with 215 separate episodes of CDI. Oral metronidazole was the most common initial treatment (145/207, 70%) followed by oral vancomycin (30/207, 15%), with recurrence rates of 30% (42/145) and 37% (11/30), respectively. Twenty-nine percent (63/215) of all initial CDI cases had at least 1 documented recurrence. Using multivariate analysis, subjects with HA-CDI were 2.6 times less likely to recur than those with CA-CDI (odds ratio: 0.39; 95% confidence interval: 0.18-0.85; P = 0.018). The overall success rate for FMT at our institution was 10/12 (83%). CONCLUSIONS Our data show that cases of HA-CDI were less likely to recur compared with CA-CDI. Although currently reserved for multiply-recurrent cases, FMT was highly successful in our small cohort. More studies on FMT should be conducted to further evaluate its usefulness in the treatment of recurrent CDI in children.
Collapse
|
473
|
Ramesh MS, Yee J. Clostridioides difficile Infection in Chronic Kidney Disease/End-Stage Renal Disease. Adv Chronic Kidney Dis 2019; 26:30-34. [PMID: 30876614 DOI: 10.1053/j.ackd.2019.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/09/2019] [Accepted: 01/09/2019] [Indexed: 11/11/2022]
Abstract
Clostridioides difficile infection (CDI) is a major health-care burden and increasingly seen in patients with chronic kidney disease (CKD) and end-stage renal disease (ESRD). Increased antibiotic use, alteration in host defenses, and gastric acid suppression are some of the etiologies for increased risk of CDI in these populations. Patients with CKD/ESRD have a higher risk of initial episode, recurrence, and development of severe CDI than those without CKD or ESRD. Diagnosis and management of CDI in patients with CKD/ESRD are similar to that in the general population. The mortality, length of stay, and health-care costs are higher in patients with CDI and CKD/ESRD. Antimicrobial stewardship with reduction in antibiotic use along with infection-control measures such as contact isolation and hand hygiene with soap and water is essential in the control and prevention of CDI in patients with CKD/ESRD.
Collapse
|
474
|
Comparison of the 2010 and 2017 Infectious Diseases Society of America guidelines on the diagnosis and treatment of Clostridium difficile infection. Curr Opin Gastroenterol 2019; 35:20-24. [PMID: 30394898 DOI: 10.1097/mog.0000000000000489] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW To highlights the key changes in the updated Infectious Diseases Society of America and Society for Healthcare Epidemiology of America guidelines with respect to the diagnosis and treatment of Clostridium difficile infection (CDI). RECENT FINDINGS CDI continues as a major threat to healthcare institutions and as a community-associated infection related primarily to antibiotic exposure. Infectious Diseases Society of America/Society for Healthcare Epidemiology of America produced extensive CDI guidelines in 2010; in 2018, updated guidance has been published. The new guidelines include key changes with respect to the treatment and diagnosis of CDI. SUMMARY Updated, evidence guidelines allow optimization of the diagnosis of CDI and the use of therapeutic interventions, in particular to reduce the risk of recurrent infection.
Collapse
|
475
|
Pouch SM, Friedman-Moraco RJ. Prevention and Treatment of Clostridium difficile-Associated Diarrhea in Solid Organ Transplant Recipients. Infect Dis Clin North Am 2018; 32:733-748. [PMID: 30146033 DOI: 10.1016/j.idc.2018.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Clostridium difficile infection is a significant cause of morbidity and mortality in solid organ transplant recipients. Risk factors in this population include frequent hospitalizations, receipt of immunosuppressive agents, and intestinal dysbiosis triggered by several factors, including exposure to broad-spectrum antimicrobials. The incidence and potential for significant adverse outcomes among solid organ transplant recipients with C difficile infection highlight the evolving need for strategic C difficile infection risk factor modification and novel approaches to disease management in this patient population. This review focuses on current concepts related to the prevention and treatment of C difficile infection in solid organ transplant recipients.
Collapse
Affiliation(s)
- Stephanie M Pouch
- Division of Infectious Diseases, Emory University School of Medicine, 101 Woodruff Circle, WMB #2101, Atlanta, GA 30322, USA.
| | - Rachel J Friedman-Moraco
- Division of Infectious Diseases, Emory University School of Medicine, 101 Woodruff Circle, WMB #2101, Atlanta, GA 30322, USA
| |
Collapse
|
476
|
Alonso CD, Mahoney MV. Bezlotoxumab for the prevention of Clostridium difficile infection: a review of current evidence and safety profile. Infect Drug Resist 2018; 12:1-9. [PMID: 30588042 PMCID: PMC6301304 DOI: 10.2147/idr.s159957] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Clostridium difficile infection (CDI) is a leading nosocomial disease estimated to cause nearly half a million cases in the United States annually. Recurrent CDI (rCDI) affects ~25% of patients after completion of standard of care therapy and is associated with substantial health care costs and a negative impact on patient's overall markers of quality of life. Bezlotoxumab is the first of its kind monoclonal antibody directed against C. difficile toxin B and indicated for prevention of rCDI in at-risk patients. For the present review, we assessed English-language studies evaluating the clinical efficacy, safety, and pharmacokinetics of bezlotoxumab in humans. Relevant studies were obtained through PubMed, Embase, Cochrane database library, Web of Science, and clinicaltrials.gov. Overall, bezlotoxumab demonstrated a 40% relative reduction rate (absolute rate reduction of ~10%) and a number needed to treat of 10 patients with a favorable safety profile. Special populations, including the elderly, immunocompromised, and patients with end-stage renal disease were evaluated in post hoc analyses with a similarly favorable reduction in rCDI. This review presents and interprets the most recent safety data and the clinical application of bezlotoxumab, highlighting specific high-risk patient populations.
Collapse
Affiliation(s)
- Carolyn D Alonso
- Department of Medicine, Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Boston, MA, USA,
- Harvard Medical School, Boston, MA, USA,
| | - Monica V Mahoney
- Department of Pharmacy, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
477
|
Tam J, Hamza T, Ma B, Chen K, Beilhartz GL, Ravel J, Feng H, Melnyk RA. Host-targeted niclosamide inhibits C. difficile virulence and prevents disease in mice without disrupting the gut microbiota. Nat Commun 2018; 9:5233. [PMID: 30531960 PMCID: PMC6286312 DOI: 10.1038/s41467-018-07705-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 11/19/2018] [Indexed: 12/13/2022] Open
Abstract
Clostridium difficile is the leading cause of nosocomial diarrhea and colitis in the industrialized world. Disruption of the protective gut microbiota by antibiotics enables colonization by multidrug-resistant C. difficile, which secrete up to three different protein toxins that are responsible for the gastrointestinal sequelae. Oral agents that inhibit the damage induced by toxins, without altering the gut microbiota, are urgently needed to prevent primary disease and break the cycle of antibiotic-induced disease recurrence. Here, we show that the anthelmintic drug, niclosamide, inhibits the pathogenesis of all three toxins by targeting a host process required for entry into colonocytes by each toxin. In mice infected with an epidemic strain of C. difficile, expressing all three toxins, niclosamide reduced both primary disease and recurrence, without disrupting the diversity or composition of the gut microbiota. Given its excellent safety profile, niclosamide may address an important unmet need in preventing C. difficile primary and recurrent diseases. Clostridium difficile causes diarrhea and colitis by producing up to three different protein toxins. Here, Tam et al. show that an anthelmintic drug, niclosamide, inhibits the pathogenesis of all three toxins by targeting a host process required for toxin entry into host cells, without disrupting the gut microbiota.
Collapse
Affiliation(s)
- John Tam
- Molecular Medicine, Hospital for Sick Children, 686 Bay St., Toronto, ON, M5G 0A4, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Therwa Hamza
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, MD, 21201, USA
| | - Bing Ma
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kevin Chen
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, MD, 21201, USA
| | - Greg L Beilhartz
- Molecular Medicine, Hospital for Sick Children, 686 Bay St., Toronto, ON, M5G 0A4, Canada
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hanping Feng
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, MD, 21201, USA
| | - Roman A Melnyk
- Molecular Medicine, Hospital for Sick Children, 686 Bay St., Toronto, ON, M5G 0A4, Canada. .,Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
478
|
Fühner V, Heine PA, Helmsing S, Goy S, Heidepriem J, Loeffler FF, Dübel S, Gerhard R, Hust M. Development of Neutralizing and Non-neutralizing Antibodies Targeting Known and Novel Epitopes of TcdB of Clostridioides difficile. Front Microbiol 2018; 9:2908. [PMID: 30574127 PMCID: PMC6291526 DOI: 10.3389/fmicb.2018.02908] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 11/13/2018] [Indexed: 12/18/2022] Open
Abstract
Clostridioides difficile is the causative bacterium in 15-20% of all antibiotic associated diarrheas. The symptoms associated with C. difficile infection (CDI) are primarily induced by the two large exotoxins TcdA and TcdB. Both toxins enter target cells by receptor-mediated endocytosis. Although different toxin receptors have been identified, it is no valid therapeutic option to prevent receptor endocytosis. Therapeutics, such as neutralizing antibodies, directly targeting both toxins are in development. Interestingly, only the anti-TcdB antibody bezlotoxumab but not the anti-TcdA antibody actoxumab prevented recurrence of CDI in clinical trials. In this work, 31 human antibody fragments against TcdB were selected by antibody phage display from the human naive antibody gene libraries HAL9/10. These antibody fragments were further characterized by in vitro neutralization assays. The epitopes of the neutralizing and non-neutralizing antibody fragments were analyzed by domain mapping, TcdB fragment phage display, and peptide arrays, to identify neutralizing and non-neutralizing epitopes. A new neutralizing epitope within the glucosyltransferase domain of TcdB was identified, providing new insights into the relevance of different toxin regions in respect of neutralization and toxicity.
Collapse
Affiliation(s)
- Viola Fühner
- Department Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Philip Alexander Heine
- Department Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Saskia Helmsing
- Department Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Sebastian Goy
- Institute for Toxicology, Hannover Medical School, Hannover, Germany
| | - Jasmin Heidepriem
- Department Synthetic Array Technologies, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Felix F. Loeffler
- Department Synthetic Array Technologies, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Stefan Dübel
- Department Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Ralf Gerhard
- Institute for Toxicology, Hannover Medical School, Hannover, Germany
| | - Michael Hust
- Department Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
479
|
Kaako A, Al-Amer M, Abdeen Y. Bezlotoxumab use as adjunctive therapy with the third fecal microbiota transplant in refractory recurrent Clostridium difficile colitis; a case report and concise literature review. Anaerobe 2018; 55:112-116. [PMID: 30521856 DOI: 10.1016/j.anaerobe.2018.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 11/26/2018] [Accepted: 11/27/2018] [Indexed: 02/08/2023]
Abstract
Clostridium difficile is the most commonly reported pathogen to cause nosocomial infections in the United States with a high burden affecting morbidity, mortality and healthcare expenditure. The use of Fecal Microbiota Transplantation (FMT) is one of the current standard therapies for recurrent C. difficile infection (CDIr). One emerging promising approach is the use of monoclonal antibodies that bind to and neutralize C. difficile toxins such as Bezlotoxumab. We present the first case report on combining the third FMT with bezlotoxumab after the failure of standard-of-care antibiotics and two trials of FMT alone, with subsequent success in preventing the recurrence of refractory CDI for 12 weeks following treatment. This case highlights the need for further studies and guidelines to recommend the best combination among different treatment options and modalities.
Collapse
Affiliation(s)
- Ahmad Kaako
- Mercy Clinic Hospitalist, Physician Building, 7301 Rogers Ave, Fort Smith, AR 72903, United States.
| | - Mohammad Al-Amer
- Internal Medicine Department, Ibn Alhaytham Hospital, Amman, Jordan
| | - Yazan Abdeen
- Pulmonary Department, Physician Building, 7301 Rogers Ave, Fort Smith, AR 72903, United States
| |
Collapse
|
480
|
Letourneau JJ, Stroke IL, Hilbert DW, Cole AG, Sturzenbecker LJ, Marinelli BA, Quintero JG, Sabalski J, Li Y, Ma L, Pechik I, Stein PD, Webb ML. Synthesis and SAR studies of novel benzodiazepinedione-based inhibitors of Clostridium difficile (C. difficile) toxin B (TcdB). Bioorg Med Chem Lett 2018; 28:3601-3605. [DOI: 10.1016/j.bmcl.2018.10.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/23/2018] [Accepted: 10/28/2018] [Indexed: 11/28/2022]
|
481
|
Management of adult Clostridium difficile digestive contaminations: a literature review. Eur J Clin Microbiol Infect Dis 2018; 38:209-231. [PMID: 30498879 DOI: 10.1007/s10096-018-3419-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 10/30/2018] [Indexed: 02/08/2023]
Abstract
Clostridium difficile infections (CDI) dramatically increased during the last decade and cause a major public health problem. Current treatments are limited by the high disease recurrence rate, severity of clinical forms, disruption of the gut microbiota, and colonization by vancomycin-resistant enterococci (VRE). In this review, we resumed current treatment options from official recommendation to promising alternatives available in the management of adult CDI, with regard to severity and recurring or non-recurring character of the infection. Vancomycin remains the first-line antibiotic in the management of mild to severe CDI. The use of metronidazole is discussed following the latest US recommendations that replaced it by fidaxomicin as first-line treatment of an initial episode of non-severe CDI. Fidaxomicin, the most recent antibiotic approved for CDI in adults, has several advantages compared to vancomycin and metronidazole, but its efficacy seems limited in cases of multiple recurrences. Innovative therapies such as fecal microbiota transplantation (FMT) and antitoxin antibodies were developed to limit the occurrence of recurrence of CDI. Research is therefore very active, and new antibiotics are being studied as surotomycin, cadazolid, and rinidazole.
Collapse
|
482
|
Basu A, Prabhu VS, Dorr MB, Golan Y, Dubberke ER, Cornely OA, Heimann SM, Pedley A, Xu R, Hanson ME, Marcella S. Bezlotoxumab Is Associated With a Reduction in Cumulative Inpatient-Days: Analysis of the Hospitalization Data From the MODIFY I and II Clinical Trials. Open Forum Infect Dis 2018; 5:ofy218. [PMID: 30460321 PMCID: PMC6237242 DOI: 10.1093/ofid/ofy218] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 10/31/2018] [Indexed: 12/17/2022] Open
Abstract
Background Patients with recurrent Clostridium difficile infection (rCDI) are more likely to have a hospital readmission and spend increased time in inpatient settings compared with patients with primary CDI. MODIFY I and II demonstrated that bezlotoxumab significantly reduced rCDI vs placebo. A post hoc within-trial analysis assessed whether bezlotoxumab was associated with a reduction in cumulative inpatient-days. Methods Data were pooled from the MODIFY trials to estimate the cumulative hospitalized days summed over the 84-day follow-up period. We adjusted inpatient use data from pooled MODIFY I and II for survival and censoring to estimate 84-day cumulative inpatient-days, overall and for subgroups. Treatment effects were obtained using recycled predictions based on trial protocol and rCDI risk, and 95% confidence intervals were obtained using 1000 bootstrap replicates. Results Mean cumulative inpatient-days were greater in the placebo arm (14.1 days) vs the bezlotoxumab arm (12.1 days) in the overall population. The mean difference between treatment groups was 2.1 days (95% confidence interval, –0.4 to –3.7). This was consistent in participants with risk factors for rCDI: age ≥65 years, compromised immunity, severe CDI, prior CDI, and ribotype 027/078/244 infection. As the number of risk factors increased, bezlotoxumab resulted in greater reductions in the number of inpatient-days compared with placebo (difference: –1.2 days, –2.3 days, –2.5 days, and –3.0 days for 0, 1, 2, and ≥3 risk factors, respectively). Conclusions Bezlotoxumab was associated with a reduction in cumulative inpatient-days, suggesting that treatment with bezlotoxumab may substantially reduce rCDI-associated health care resource use. Trial registrations. MODIFY I (MK-3415A-001, NCT01241552) and II (MK-3415A-002, NCT01513239)
Collapse
Affiliation(s)
- Anirban Basu
- The Comparative Health Outcomes, Policy, and Economics (CHOICE) Institute, University of Washington, Seattle, Washington
| | | | | | - Yoav Golan
- New England Medical Center, Tufts University, Boston, Massachusetts
| | - Erik R Dubberke
- Department of Medicine, Washington University, St. Louis, Missouri
| | - Oliver A Cornely
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Department I of Internal Medicine, ECMM Excellence Center of Medical Mycology, Clinical Trials Centre Cologne (ZKS Köln), University of Cologne, Cologne, Germany
| | - Sebastian M Heimann
- Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | | | - Ruifeng Xu
- Merck & Co., Inc., Kenilworth, New Jersey
| | | | | |
Collapse
|
483
|
Comparing fecal microbiota transplantation to standard-of-care treatment for recurrent Clostridium difficile infection: a systematic review and meta-analysis. Eur J Gastroenterol Hepatol 2018; 30:1309-1317. [PMID: 30138161 DOI: 10.1097/meg.0000000000001243] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The use of fecal microbiota transplantation (FMT) as a treatment option for recurrent Clostridium difficile infection (rCDI) is well established. Various studies have used different forms and administration routes for FMT. We performed a systemic review and meta-analysis to update the clinical knowledge about different FMT modalities for curing rCDI compared with medical treatment (MT). PATIENTS AND METHODS We searched PubMed and Medline from inception through 10 May 2018 for randomized control trials (RCTs) comparing FMT (fresh or frozen) versus MT. We used Cochrane Collaboration's Risk of Bias tool to assess bias in the RCTs. We estimated odds ratio (OR) with 95% confidence interval (CI) for each outcome using a random effects model. P values of less than 0.05 were considered significant. RESULTS We included seven RCTs comprising a total of 543 patients with recurrent CDI. There was a nonsignificant trend toward resolution of diarrhea following a single fresh FMT infusion compared with frozen FMT and MT (OR=2.45, 95% CI=0.78-7.71, P=0.12, I=69%). Subgroup analysis of fresh FMT vs. frozen FMT showed no difference between the two groups (OR=2.13, 95% CI=0.22-20.41, P=0.51, I=61%). Frozen FMT infusion through upper route versus lower route showed no difference (OR=0.62, 95% CI=0.15-2.54, P=0.51, I=0%). There was a nonsignificant trend favoring multiple treatments with FMT versus multiple courses of MT (OR=3.68, 95% CI=0.74-18.22, P=0.11, I=0%). CONCLUSION FMT is a promising treatment modality for rCDI compared with MT alone. Different forms and routes of FMT administration seem to be equally efficacious. In future, more well-designed RCTs directed at homogenous FMT preparation and delivery methods are required to validate these findings.
Collapse
|
484
|
Salavert M, Cobo J, Pascual Á, Aragón B, Maratia S, Jiang Y, Aceituno S, Grau S. Cost-Effectiveness Analysis of Bezlotoxumab Added to Standard of Care Versus Standard of Care Alone for the Prevention of Recurrent Clostridium difficile Infection in High-Risk Patients in Spain. Adv Ther 2018; 35:1920-1934. [PMID: 30328061 PMCID: PMC6223985 DOI: 10.1007/s12325-018-0813-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Clostridium difficile infection (CDI) is the major cause of infectious nosocomial diarrhoea and is associated with considerable morbidity, mortality and economic impact. Bezlotoxumab administered in combination with standard of care (SoC) antibiotic therapy prevents recurrent CDI. This study assessed the cost-effectiveness of bezlotoxumab added to SoC, compared to SoC alone, to prevent the recurrence of CDI in high-risk patients from the Spanish National Health System perspective. METHODS A Markov model was used to simulate the natural history of CDI over a lifetime horizon in five populations of patients at high risk of CDI recurrence according to MODIFY trials: (1) ≥ 65 years old; (2) severe CDI; (3) immunocompromised; (4) ≥ 1 CDI episode in the previous 6 months; and (5) ≥ 65 years old and with ≥ 1 CDI episode in the previous 6 months. The incremental cost-effectiveness ratio (ICER) expressed as cost per quality-adjusted life-year (QALY) gained was calculated. Deterministic (DSA) and probabilistic sensitivity analyses (PSA) were performed. RESULTS In all patient populations (from 1 to 5), bezlotoxumab added to SoC reduced CDI recurrence compared to SoC alone by 26.4, 19.5, 21.2, 26.6 and 39.7%, respectively. The resulting ICERs for the respective subgroups were €12,724, €17,495, €9545, €7386, and €4378. The model parameters with highest impact on the ICER were recurrence rate (first), mortality, and utility values. The probability that bezlotoxumab was cost-effective at a willingness-to-pay threshold of €21,000/QALY was 85.5%, 54.1%, 86.0%, 94.5%, 99.6%, respectively. CONCLUSION The results suggest that bezlotoxumab added to SoC compared to SoC alone is a cost-effective treatment to prevent the recurrence of CDI in high-risk patients. The influence of changes in model parameters on DSA results was higher in patients ≥ 65 years old, with severe CDI and immunocompromised. Additionally, PSA estimated that the probability of cost-effectiveness exceeded 85% in most subgroups. FUNDING Merck Sharp & Dohme Corp.
Collapse
Affiliation(s)
- Miguel Salavert
- Infectious Diseases Unit, Hospital Universitario La Fe, Valencia, Spain.
| | - Javier Cobo
- Infectious Diseases Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Álvaro Pascual
- Clinical Microbiology and Infectious Diseases Unit, Hospital Universitario Virgen Macarena, Instituto de Biomedicina de Sevilla (IBIS), Universidad de Sevilla, Seville, Spain
| | | | | | - Yiling Jiang
- Merck Sharp & Dohme Ltd, Hoddesdon, Hertfordshire, UK
| | | | - Santiago Grau
- Pharmacy Department, Hospital del Mar, Universidad Autónoma de Barcelona, Barcelona, Spain
| |
Collapse
|
485
|
Igarashi Y, Tashiro S, Enoki Y, Taguchi K, Matsumoto K, Ohge H, Suzuki H, Nakamura A, Mori N, Morinaga Y, Yamagishi Y, Yoshizawa S, Yanagihara K, Mikamo H, Kunishima H. Oral vancomycin versus metronidazole for the treatment of Clostridioides difficile infection: Meta-analysis of randomized controlled trials. J Infect Chemother 2018; 24:907-914. [PMID: 30170735 DOI: 10.1016/j.jiac.2018.08.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 07/27/2018] [Accepted: 08/03/2018] [Indexed: 12/17/2022]
Abstract
At present, vancomycin (VCM) and metronidazole (MNZ) are used for the first-line standard treatment of Clostridioides difficile infection (CDI). However, their differential use has not been sufficiently investigated. In this study, a meta-analysis on differences in the efficacy for CDI between VCM and MNZ was performed. Reports of randomized controlled studies using VCM or MNZ to treat CDI were surveyed. Meta-analysis was performed using the Mantel-Haenszel method and random-effects model, and the risk ratio and 95% confidence interval were calculated. Excluding overlapping reports, 1043 reports were extracted and 5 randomized controlled studies were extracted. There was no difference in therapeutic effects for CDI between VCM and MNZ (RR = 1.08, 95% CI (0.99-1.17), p = 0.09, I2 = 37%). On subgroup analysis by the severity, there was no difference in the clinical effects for CDI between VCM and MNZ in non-severe cases (risk ratio: 1.09, 95% confidence interval: 1.00-1.19, p = 0.06), but the clinical effects of VCM were significantly higher than those of MNZ in severe cases (risk ratio: 1.19, 95% confidence interval: 1.02-1.39, p = 0.03). No significant difference was noted in the recurrence rate, incidence of adverse event, time to exhibit therapeutic effects, or judgment of the bacteriological effects. As the therapeutic effects of VCM were superior in severe CDI cases, VCM should be considered first in severe cases.
Collapse
Affiliation(s)
- Yuki Igarashi
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, Tokyo, Japan
| | - Sho Tashiro
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, Tokyo, Japan
| | - Yuki Enoki
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, Tokyo, Japan.
| | - Kazuaki Taguchi
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, Tokyo, Japan
| | - Kazuaki Matsumoto
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, Tokyo, Japan
| | - Hiroki Ohge
- Department of Infectious Diseases, Hiroshima University Hospital, Hiroshima, Japan
| | - Hiromichi Suzuki
- Division of Infectious Diseases, Department of Medicine, Tsukuba Medical Center Hospital, Ibaraki, Japan
| | - Atsushi Nakamura
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| | - Nobuaki Mori
- Department of General Internal Medicine, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Yoshitomo Morinaga
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yuka Yamagishi
- Department of Clinical Infectious Diseases, Aichi Medical University Hospital, Aichi, Japan; Department of Infection Control and Prevention, Aichi Medical University Hospital, Aichi, Japan
| | - Sadako Yoshizawa
- Clinical Research Center, Department of Microbiology and Infectious Diseases, Toho University Faculty of Medicine, Tokyo, Japan
| | - Katsunori Yanagihara
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hiroshige Mikamo
- Department of Clinical Infectious Diseases, Aichi Medical University Hospital, Aichi, Japan; Department of Infection Control and Prevention, Aichi Medical University Hospital, Aichi, Japan
| | - Hiroyuki Kunishima
- Department of Infectious Diseases, St. Marianna University, Kanagawa, Japan
| |
Collapse
|
486
|
Mullish BH, Quraishi MN, Segal JP, McCune VL, Baxter M, Marsden GL, Moore DJ, Colville A, Bhala N, Iqbal TH, Settle C, Kontkowski G, Hart AL, Hawkey PM, Goldenberg SD, Williams HRT. The use of faecal microbiota transplant as treatment for recurrent or refractory Clostridium difficile infection and other potential indications: joint British Society of Gastroenterology (BSG) and Healthcare Infection Society (HIS) guidelines. Gut 2018; 67:1920-1941. [PMID: 30154172 DOI: 10.1136/gutjnl-2018-316818] [Citation(s) in RCA: 233] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/27/2018] [Accepted: 07/01/2018] [Indexed: 12/16/2022]
Abstract
Interest in the therapeutic potential of faecal microbiota transplant (FMT) has been increasing globally in recent years, particularly as a result of randomised studies in which it has been used as an intervention. The main focus of these studies has been the treatment of recurrent or refractory Clostridium difficile infection (CDI), but there is also an emerging evidence base regarding potential applications in non-CDI settings. The key clinical stakeholders for the provision and governance of FMT services in the UK have tended to be in two major specialty areas: gastroenterology and microbiology/infectious diseases. While the National Institute for Health and Care Excellence (NICE) guidance (2014) for use of FMT for recurrent or refractory CDI has become accepted in the UK, clear evidence-based UK guidelines for FMT have been lacking. This resulted in discussions between the British Society of Gastroenterology (BSG) and Healthcare Infection Society (HIS), and a joint BSG/HIS FMT working group was established. This guideline document is the culmination of that joint dialogue.
Collapse
Affiliation(s)
- Benjamin H Mullish
- Division of Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.,Departments of Gastroenterology and Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Mohammed Nabil Quraishi
- Department of Gastroenterology, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Jonathan P Segal
- Division of Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.,Inflammatory Bowel Disease Unit, St Mark's Hospital, London, UK
| | - Victoria L McCune
- Public Health England, Public Health Laboratory Birmingham, Birmingham, UK.,Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Melissa Baxter
- Department of Microbiology, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | | | - David J Moore
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Alaric Colville
- Department of Microbiology, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Neeraj Bhala
- Department of Gastroenterology, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Institute of Applied Health Research, University of Birmingham, Birmingham, UK.,Institute of Translational Medicine, University of Birmingham, Birmingham, UK
| | - Tariq H Iqbal
- Department of Gastroenterology, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Institute of Translational Medicine, University of Birmingham, Birmingham, UK
| | - Christopher Settle
- Department of Microbiology, City Hospitals Sunderland NHS Foundation Trust, Sunderland, Sunderland, UK
| | | | - Ailsa L Hart
- Division of Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.,Inflammatory Bowel Disease Unit, St Mark's Hospital, London, UK
| | - Peter M Hawkey
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Simon D Goldenberg
- Centre for Clinical Infection and Diagnostics Research, King's College London, London, UK.,Department of Microbiology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Horace R T Williams
- Division of Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.,Departments of Gastroenterology and Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
487
|
Diorio C, Robinson PD, Ammann RA, Castagnola E, Erickson K, Esbenshade A, Fisher BT, Haeusler GM, Kuczynski S, Lehrnbecher T, Phillips R, Cabral S, Dupuis LL, Sung L. Guideline for the Management of Clostridium Difficile Infection in Children and Adolescents With Cancer and Pediatric Hematopoietic Stem-Cell Transplantation Recipients. J Clin Oncol 2018; 36:3162-3171. [PMID: 30216124 PMCID: PMC6209092 DOI: 10.1200/jco.18.00407] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The aim of this work was to develop a clinical practice guideline for the prevention and treatment of Clostridium difficile infection (CDI) in children and adolescents with cancer and pediatric hematopoietic stem-cell transplantation (HSCT) patients. METHODS An international multidisciplinary panel of experts in pediatric oncology and infectious diseases with patient advocate representation was convened. We performed systematic reviews of randomized controlled trials for the prevention or treatment of CDI in any population and considered the directness of the evidence to children with cancer and pediatric HSCT patients. We used the Grading of Recommendations Assessment, Development, and Evaluation approach to generate recommendations. RESULTS The panel made strong recommendations to administer either oral metronidazole or oral vancomycin for the initial treatment of nonsevere CDI and oral vancomycin for the initial treatment of severe CDI. Fidaxomicin may be considered in the setting of recurrent CDI. The panel suggested that probiotics not be routinely used for the prevention of CDI, and that monoclonal antibodies and probiotics not be routinely used for the treatment of CDI. A strong recommendation to not use fecal microbiota transplantation was made in this population. We identified key knowledge gaps and suggested directions for future research. CONCLUSION We present a guideline for the prevention and treatment of CDI in children and adolescents with cancer and pediatric HSCT patients. Future research should include randomized controlled trials that involve children with cancer and pediatric HSCT patients to improve the management of CDI in this population.
Collapse
Affiliation(s)
- Caroline Diorio
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Paula D. Robinson
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Roland A. Ammann
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Elio Castagnola
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Kelley Erickson
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Adam Esbenshade
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Brian T. Fisher
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Gabrielle M. Haeusler
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Susan Kuczynski
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Thomas Lehrnbecher
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Robert Phillips
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Sandra Cabral
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - L. Lee Dupuis
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Lillian Sung
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom.,Corresponding author: Lillian Sung, MD, PhD, Division of Haematology/Oncology, The Hospital for Sick Children, 555 University Ave, Toronto, ON M5G1X8, Canada; e-mail:
| |
Collapse
|
488
|
Chung SY, Schöttelndreier D, Tatge H, Fühner V, Hust M, Beer LA, Gerhard R. The Conserved Cys-2232 in Clostridioides difficile Toxin B Modulates Receptor Binding. Front Microbiol 2018; 9:2314. [PMID: 30416488 PMCID: PMC6212469 DOI: 10.3389/fmicb.2018.02314] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/11/2018] [Indexed: 12/14/2022] Open
Abstract
Clostridioides difficile toxins TcdA and TcdB are large clostridial glucosyltransferases which are the main pathogenicity factors in C. difficile-associated diseases. Four highly conserved cysteines are present in all large clostridial glucosyltransferases. In this study we focused on the conserved cysteine 2232 within the combined repetitive oligopeptide domain of TcdB from reference strain VPI10463 (clade I). Cysteine 2232 is not present in TcdB from hypervirulent strain R20291 (clade II), where a tyrosine is found instead. Replacement of cysteine 2232 by tyrosine in TcdBV PI10463 reduced binding to the soluble fragments of the two known TcdB receptors, frizzled-2 (FZD2) and poliovirus receptor-like protein-3/nectin-3 (PVRL3). In line with this, TcdBR20291 showed weak binding to PVRL3 in pull-down assays which was increased when tyrosine 2232 was exchanged for cysteine. Surprisingly, we did not observe binding of TcdBR20291 to FZD2, indicating that this receptor is less important for this toxinotype. Competition assay with the receptor binding fragments (aa 1101–1836) of TcdBV PI10463 and TcdBR20291, as well as antibodies newly developed by antibody phage display, revealed different characteristics of the yet poorly described delivery domain of TcdB harboring the second receptor binding region. In summary, we found that conserved Cys-2232 in TcdB indirectly contributes to toxin–receptor interaction.
Collapse
Affiliation(s)
- Soo-Young Chung
- Institute of Toxicology, Hannover Medical School, Hanover, Germany
| | | | - Helma Tatge
- Institute of Toxicology, Hannover Medical School, Hanover, Germany
| | - Viola Fühner
- Department of Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Michael Hust
- Department of Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | | | - Ralf Gerhard
- Institute of Toxicology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
489
|
Mejia-Chew C, Dubberke ER. Clostridium difficile control measures: current and future methods for prevention. Expert Rev Anti Infect Ther 2018; 16:121-131. [PMID: 29353504 DOI: 10.1080/14787210.2018.1429911] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Clostridium difficile is the most common cause of healthcare associated infection, and C. difficile infection (CDI) is associated with significant costs, morbidity, and mortality. One obstacle to preventing CDI is lack of high quality data on interventions to prevent CDI. This has led some to focus on areas, such as method of hand hygiene, unlikely to impact CDI incidence as much as others, such as contact precautions. In addition, existing strategies, although effective, do have limitations. Another challenge is the ability to rapidly, and accurately, diagnose CDI. Given these obstacles, new strategies to effectively prevent CDI are imperative to improve patient outcomes. Areas covered: Evidence of the interventions recommended by international scientific societies will be reviewed, as well as ongoing research on new strategies, such as screening for asymptomatic C. difficile carriage, microbiota sparing agents, bacteriocins and vaccines. Expert commentary: Current measures to prevent CDI are effective, but have significant limitations. Contact precautions and antimicrobial stewardship are likely the most effective of current prevention recommendations. Diagnostic assay utilization plays a role as well. New strategies to prevent CDI are needed, and, fortunately, several are being studied. Most likely a combination of approaches will be necessary to optimize CDI prevention.
Collapse
Affiliation(s)
- Carlos Mejia-Chew
- a Division of Infectious Disease , Washington University School of Medicine , St Louis , MO , USA
| | - Erik R Dubberke
- a Division of Infectious Disease , Washington University School of Medicine , St Louis , MO , USA
| |
Collapse
|
490
|
Abughanimeh O, Qasrawi A, Kaddourah O, Al Momani L, Abu Ghanimeh M. Clostridium difficile infection in oncology patients: epidemiology, pathophysiology, risk factors, diagnosis, and treatment. Hosp Pract (1995) 2018; 46:266-277. [PMID: 30296190 DOI: 10.1080/21548331.2018.1533673] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Clostridium difficile infection (CDI) is one of the most common healthcare-associated infections in the United States. Its incidence has been increasing in the recent years despite preventative measures. CDI increases annual expenses by 1.5 billion dollars. Cancer patients are at higher risk to acquire CDI, as explained by their frequent exposure to risk factors. CDI in cancer patients is associated with higher mortality rates and prolonged hospitalization. Furthermore, CDI affects the course of the disease by delaying treatments such as chemotherapy. Chemotherapeutics drugs are considered independent risk factors for CDI. This review discusses Clostridium difficile infection in cancer patients, including those who are receiving chemotherapy. Herein, we summarize recent data regarding the epidemiology, risk factors, including chemotherapy regimens, pathogenesis, diagnostic techniques and treatment options, including newer agents. Method: A literature search was performed using the PubMed and Google Scholar databases. The MeSH terms utilized in different combinations were 'clostridium difficile', 'neoplasia/cancer/oncology', 'chemotherapy', 'diagnosis', and 'treatment', in addition to looking up each treatment option individually to generate a comprehensive search. The articles were initially screened by title alone, followed by screening through abstracts. Full texts of pertinent articles (including letters to editors, case reports, case series, cohort studies, and clinical trials) were included in this review.
Collapse
Affiliation(s)
- Omar Abughanimeh
- a School of Medicine Internal Medicine , University of Missouri , Kansas City , USA
| | - Ayman Qasrawi
- a School of Medicine Internal Medicine , University of Missouri , Kansas City , USA
| | - Osama Kaddourah
- a School of Medicine Internal Medicine , University of Missouri , Kansas City , USA
| | - Laith Al Momani
- b East Tennessee State University James H Quillen College of Medicine - Internal Medicine , USA
| | | |
Collapse
|
491
|
Calvert MB, Jumde VR, Titz A. Pathoblockers or antivirulence drugs as a new option for the treatment of bacterial infections. Beilstein J Org Chem 2018; 14:2607-2617. [PMID: 30410623 PMCID: PMC6204809 DOI: 10.3762/bjoc.14.239] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/20/2018] [Indexed: 12/19/2022] Open
Abstract
The rapid development of antimicrobial resistance is threatening mankind to such an extent that the World Health Organization expects more deaths from infections than from cancer in 2050 if current trends continue. To avoid this scenario, new classes of anti-infectives must urgently be developed. Antibiotics with new modes of action are needed, but other concepts are also currently being pursued. Targeting bacterial virulence as a means of blocking pathogenicity is a promising new strategy for disarming pathogens. Furthermore, it is believed that this new approach is less susceptible towards resistance development. In this review, recent examples of anti-infective compounds acting on several types of bacterial targets, e.g., adhesins, toxins and bacterial communication, are described.
Collapse
Affiliation(s)
- Matthew B Calvert
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Varsha R Jumde
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Alexander Titz
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany.,Department of Pharmacy, Saarland University, Saarbrücken, Germany
| |
Collapse
|
492
|
Paknikar R, Pekow J. Fecal Microbiota Transplantation for the Management of Clostridium difficile Infection. Surg Infect (Larchmt) 2018; 19:785-791. [PMID: 30300561 DOI: 10.1089/sur.2018.221] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The clinical burden of Clostridium difficile infection (CDI) continues to grow. Despite the multitude of treatment options that have been developed and tested to combat the morbidity and death associated with CDI, recurrence remains common. As such, treatment modalities such as fecal microbiota transplantation (FMT) have become studied increasingly; FMT serves to transplant stool from carefully selected healthy subjects into C. difficile positive patients through a variety of delivery routes. In doing so, FMT is hypothesized to correct dysbiosis of the recipient gut microbiome addressing the root cause of the pathogenesis of C. difficile infection. A growing body of evidence shows FMT to be efficacious in this setting, and the study of FMT accordingly continues to evolve to identify novel indications for its utilization.
Collapse
Affiliation(s)
- Raghavendra Paknikar
- Section of Gastroenterology, Hepatology, and Nutrition, University of Chicago , Chicago, Illinois
| | - Joel Pekow
- Section of Gastroenterology, Hepatology, and Nutrition, University of Chicago , Chicago, Illinois
| |
Collapse
|
493
|
Abstract
Clostridioides difficile (formerly Clostridium difficile) infection is the most frequently identified health care-associated infection in the United States. C difficile has also emerged as a cause of community-associated diarrhea, resulting in increased incidence of community-associated infection. Clinical illness ranges in severity from mild diarrhea to fulminant colitis and death. Appropriate management of infection requires understanding of the various diagnostic assays and therapeutic options as well as relevant measures to infection prevention. This article provides updated recommendations regarding the prevention, diagnosis, and treatment of incident and recurrent C difficile infection.
Collapse
Affiliation(s)
- Alice Y Guh
- From the Centers for Disease Control and Prevention, Atlanta, Georgia. (A.Y.G., P.K.K.)
| | - Preeta K Kutty
- From the Centers for Disease Control and Prevention, Atlanta, Georgia. (A.Y.G., P.K.K.)
| |
Collapse
|
494
|
Kelly CP, Wilcox MH, Glerup H, Aboo N, Ellison MC, Eves K, Dorr MB. Bezlotoxumab for Clostridium difficile Infection Complicating Inflammatory Bowel Disease. Gastroenterology 2018; 155:1270-1271. [PMID: 30227108 DOI: 10.1053/j.gastro.2018.06.080] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/11/2018] [Accepted: 06/18/2018] [Indexed: 12/02/2022]
Affiliation(s)
| | - Mark H Wilcox
- Leeds Teaching Hospitals and University of Leeds, Leeds, UK
| | | | | | | | | | | |
Collapse
|
495
|
Dubberke ER, Lee CH, Orenstein R, Khanna S, Hecht G, Gerding DN. Results From a Randomized, Placebo-Controlled Clinical Trial of a RBX2660-A Microbiota-Based Drug for the Prevention of Recurrent Clostridium difficile Infection. Clin Infect Dis 2018; 67:1198-1204. [PMID: 29617739 DOI: 10.1093/cid/ciy259] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/28/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Despite advancements, recurrent Clostridium difficile infections (CDI) remain an urgent public health threat with insufficient response rates to currently approved antibiotic therapies. Microbiota-based treatments appear effective, but rigorous clinical trials are required to optimize dosing strategies and substantiate long-term safety. METHODS This randomized, double-blind, placebo-controlled phase 2B trial enrolled adults with 2 or more CDI recurrences to receive: 2 doses of RBX2660, a standardized microbiota-based drug (group A); 2doses of placebo (group B); or 1 dose of RBX2660 followed by 1 dose of placebo (group C). Efficacy was defined as prevention of recurrent CDI for 8 weeks following treatment. Participants who had a recurrence within 8 weeks were eligible to receive up to 2 open-label RBX2660 doses. The primary endpoint was efficacy for group A compared to group B. Secondary endpoints included the efficacy of group C compared to group B, combined efficacy in the blinded and open-label phases, and safety for 24 months. RESULTS The efficacy for groups A, B, and C were 61%, 45%, and 67%, respectively. The primary endpoint was not met (P = .152). One RBX2660 dose (group C) was superior to placebo (group B; P = .048), and the overall efficacy (including open-label response) for RBX2660-treated participants was 88.8%. Adverse events did not differ significantly among treatment groups. CONCLUSIONS One, but not 2, doses of RBX2660 was superior to placebo in this randomized, placebo-controlled trial. These data provide important insights for a larger phase 3 trial and continued clinical development of RBX2660. CLINICAL TRIALS REGISTRATION NCT02299570.
Collapse
Affiliation(s)
- Erik R Dubberke
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Christine H Lee
- Hamilton Regional Laboratory Medicine Program, Department of Pathology and Molecular Medicine, McMaster University, Ontario
- Vancouver Island Health Authority, Victoria, British Columbia
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Robert Orenstein
- Division of Infectious Diseases, Mayo Clinic in Arizona, Phoenix
| | - Sahil Khanna
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Gail Hecht
- Division of Gastroenterology, Hepatology and Nutrition, Loyola University Medical Center, Chicago, Illinois
| | | |
Collapse
|
496
|
Daniels LM, Kufel WD. Clinical review of Clostridium difficile infection: an update on treatment and prevention. Expert Opin Pharmacother 2018; 19:1759-1769. [PMID: 30220230 DOI: 10.1080/14656566.2018.1524872] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Clostridium difficile infection (CDI) has become a significant healthcare-associated infection and is strongly associated with antibiotic use. Practice guidelines have recently been revised incorporating updated recommendations for diagnosis, treatment, and prevention. AREAS COVERED This review discusses updated aspects of CDI management. New and emerging pharmacologic options for treatment and prevention are reviewed. EXPERT OPINION Metronidazole is associated with lower rates of treatment success compared to vancomycin and should no longer be used as primary therapy for the first episode of CDI or recurrent disease. Vancomycin or fidaxomicin are now recommended for first-line therapy for most cases of CDI. Fecal microbiota transplant is effective and safe for the treatment of recurrent CDI. Evidence supports the use of fidaxomicin and bezlotoxumab for prevention of recurrent CDI; however, the costs associated with these therapies may limit their use. Validated risk prediction tools are needed to identify patients most likely to benefit from these treatments. Future advancements in microbiota targeting treatments will emerge as promising alternatives to standard CDI treatments. Antibiotic stewardship and infection control measures will remain essential components for CDI management.
Collapse
Affiliation(s)
- Lindsay M Daniels
- a Department of Pharmacy , University of North Carolina Medical Center , Chapel Hill , NC , USA.,b Division of Practice Advancement and Clinical Education, Eshelman School of Pharmacy , University of North Carolina , Chapel Hill , NC , USA
| | - Wesley D Kufel
- c Department of Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences , Binghamton University , Binghamton , NY , USA.,d Department of Medicine , Upstate Medical University.,e Department of Pharmacy , Upstate University Hospital , Syracuse , NY , USA
| |
Collapse
|
497
|
Grześkowiak ŁM, Pieper R, Huynh HA, Cutting SM, Vahjen W, Zentek J. Impact of early-life events on the susceptibility to Clostridium difficile colonisation and infection in the offspring of the pig. Gut Microbes 2018; 10:251-259. [PMID: 30252612 PMCID: PMC6546313 DOI: 10.1080/19490976.2018.1518554] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Clostridium difficile has been documented as a major cause of uncontrolled outbreaks of enteritis in neonatal pigs and antibiotic-associated infections in clinical settings. It belongs to the natural cohort of early colonisers of the gastrointestinal tract of pigs and can be detected in faeces up to two weeks post-partum. In older pigs, it often remains under the detection limit. Most neonatal pigs show no clinical signs of disease although C. difficile and its toxins can be detected at high levels in faeces. Increased mortality rates associated with C. difficile on pig farms are, so far, considered "spontaneous" and the predisposing factors are mostly not defined. The infection caused by C. difficile is multifactorial and it is likely that the repertoire of maternal factors, host physiology, the individually developing gut microbiota, co-infections and environmental stress define the conditions for disease development. In this addendum to our recently published work on CDI in neonatal piglets, we discuss the "early-life events" that influence C. difficile spread and infection in neonatal piglets.
Collapse
Affiliation(s)
- Łukasz M. Grześkowiak
- Institute of Animal Nutrition, Freie Universität Berlin, Berlin, Germany,CONTACT Łukasz M. Grześkowiak
| | - Robert Pieper
- Institute of Animal Nutrition, Freie Universität Berlin, Berlin, Germany
| | - Hong A. Huynh
- School of Biological Sciences, Royal Holloway University of London, Egham, UK
| | - Simon M. Cutting
- School of Biological Sciences, Royal Holloway University of London, Egham, UK
| | - Wilfried Vahjen
- Institute of Animal Nutrition, Freie Universität Berlin, Berlin, Germany
| | - Jürgen Zentek
- Institute of Animal Nutrition, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
498
|
Beinortas T, Burr NE, Wilcox MH, Subramanian V. Comparative efficacy of treatments for Clostridium difficile infection: a systematic review and network meta-analysis. THE LANCET. INFECTIOUS DISEASES 2018; 18:1035-1044. [DOI: 10.1016/s1473-3099(18)30285-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/06/2018] [Accepted: 04/24/2018] [Indexed: 12/11/2022]
|
499
|
Cobo J. A comprehensive approach for the patient with Clostridium difficile infection. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2018; 31 Suppl 1:27-31. [PMID: 30209919 PMCID: PMC6459568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
During the last decade there have been many changes and advances in the research on Clostridium difficile infection (CDI). We have improved diagnostic and therapeutic tools and, at the same time, we have learned that the CDI implies, especially in the most vulnerable patients, an important morbidity. CDI has traditionally been undervalued and it is widely dispersed in hospitals. Surely, there is inertness in its management and there are also broad areas of improvement. If we add to this the high cost of the new drugs and the practical difficulties to implement the faecal microbiota transplant, we realize that we may not be taking full advantage of all the opportunities to improve patient's outcomes. The implementation of policies that favour the supervision of all CDI cases by an expert in infectious diseases will contribute to a better global management of this important disease.
Collapse
|
500
|
Tampaki EC, Tampakis A, Posabella A, Patsouris E, Kontzoglou K, Kouraklis G. Current clostridium difficile treatments: Lessons that need to be learned from the clinical trials. Hum Vaccin Immunother 2018; 14:2874-2875. [PMID: 30148975 DOI: 10.1080/21645515.2018.1493327] [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: 10/28/2022] Open
Abstract
Clostridium difficile infection (CDI) is the most common infectious disease cause of nosocomial diarrhea in adults in developed countries. Judging from the clinical trials on drugs used in CDIs, no approved treatment for recurrences exists, possibly indicating that a combination of treatment approaches are mandatory especially in severe infections, with current studies not being fully representative. Among the new strategies researched intensively fidaxomicin is presented, which demonstrates reduced CDI recurrences. Moreover, biotherapeutic strategies, mainly fecal microbiota transplantation but also competitive inhibition with non-toxigenic strains of C. difficile, and finally monoclonal antibodies against C. difficile toxins which offer protection against recurrences. Careful interpretation of the results based on lessons learned from previous trials conducted seems crucial. Questions are raised regarding how the results of future studies regarding new strategies researched will be managed and interpreted especially with regard to recurrence management as relevant data must be monitored for at least 30 days after end of treatment.
Collapse
Affiliation(s)
- E C Tampaki
- a Department of Propaedeutic Surgery , Laikon General Hospital, School of Medicine, National and Kapodistrian University of Athens , Greece.,b National Organization for the Provision of Healthcare Services , Department of Planning and Monitoring of Medicines Dispensing, Medicines Division , Greece
| | - A Tampakis
- a Department of Propaedeutic Surgery , Laikon General Hospital, School of Medicine, National and Kapodistrian University of Athens , Greece.,c Department of General and Visceral Surgery , University Hospital of Basel , Switzerland
| | - A Posabella
- d Department of Visceral Surgery , St. Clara Hospital , Basel , Switzerland
| | - E Patsouris
- e Department of Pathology , School of Medicine, National and Kapodistrian University of Athens , Greece
| | - K Kontzoglou
- a Department of Propaedeutic Surgery , Laikon General Hospital, School of Medicine, National and Kapodistrian University of Athens , Greece
| | - G Kouraklis
- a Department of Propaedeutic Surgery , Laikon General Hospital, School of Medicine, National and Kapodistrian University of Athens , Greece
| |
Collapse
|