151
|
Wu Q, Savidge TC. Systems approaches for the clinical diagnosis of Clostridioides difficile infection. Transl Res 2020; 220:57-67. [PMID: 32272094 DOI: 10.1016/j.trsl.2020.03.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/02/2020] [Accepted: 03/09/2020] [Indexed: 12/18/2022]
Abstract
Clostridioides difficile infection (CDI) is an urgent threat to global public health. Patient susceptibility to C. difficile is highly dependent on host immune status and gut dysbiosis resulting in loss of protective microbiota consortia that prevent spore germination, pathogen colonization, and disease pathogenesis. Recent clinical studies highlight the problems of differentiating symptomatic CDI from asymptomatic C. difficile carriage in patients with diarrhea. In this review, we consider how integration of microbiome and host immune systems biology data may aid in the clinical diagnosis of CDI when validated against gold standard testing and combined with standard microbiology laboratory assays.
Collapse
Affiliation(s)
- Qinglong Wu
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Texas Children's Microbiome Center, Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Tor C Savidge
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Texas Children's Microbiome Center, Department of Pathology, Texas Children's Hospital, Houston, Texas.
| |
Collapse
|
152
|
Scaccia N, Vaz-Moreira I, Manaia CM. Persistence of wastewater antibiotic resistant bacteria and their genes in human fecal material. FEMS Microbiol Ecol 2020; 96:5815073. [PMID: 32239211 DOI: 10.1093/femsec/fiaa058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 03/27/2020] [Indexed: 01/11/2023] Open
Abstract
Domestic wastewater is a recognized source of antibiotic resistant bacteria and antibiotic resistance genes (ARB&ARGs), whose risk of transmission to humans cannot be ignored. The fitness of wastewater ARB in the complex fecal microbiota of a healthy human was investigated in feces-based microcosm assays (FMAs). FMAs were inoculated with two wastewater isolates, Escherichia coli strain A2FCC14 (MLST ST131) and Enterococcus faecium strain H1EV10 (MLST ST78), harboring the ARGs blaTEM, blaCTX, blaOXA-A and vanA, respectively. The FMAs, incubated in the presence or absence of oxygen or in the presence or absence of the antibiotics cefotaxime or vancomycin, were monitored based on cultivation, ARGs quantification and bacterial community analysis. The fecal bacterial community was dominated by members of the phyla Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria and Verrucomicrobia. The ARGs harbored by the wastewater isolates could be quantified after one week, in FMAs incubated under both aerobic and anaerobic conditions. These observations were not significantly different in FMAs incubated anaerobically, supplemented with sub-inhibitory concentrations of cefotaxime or vancomycin. The observation that ARGs of wastewater ARB persisted in presence of the human fecal microbiota for at least one week supports the hypothesis of a potential transmission to humans, a topic that deserves further investigation.
Collapse
Affiliation(s)
- Nazareno Scaccia
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Ivone Vaz-Moreira
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Célia M Manaia
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| |
Collapse
|
153
|
Li G, Xia X, Zhao S, Shi M, Liu F, Zhu Y. The physiological and toxicological effects of antibiotics on an interspecies insect model. CHEMOSPHERE 2020; 248:126019. [PMID: 32007775 DOI: 10.1016/j.chemosphere.2020.126019] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 06/10/2023]
Abstract
Silkworm (Bombyx mori L.) has a clear genetic background, parts of which are highly homologous to certain genes related to human hereditary diseases. Thus, the species presents an excellent interspecies model for drug screening and microbe-host interaction studies. Chloramphenicol (CAM) and vancomycin (VCM) are antibiotics commonly used to treat specific bacterial infections in medical care, animal husbandry, and agriculture. However, inappropriate dosages and prolonged therapy increase their risk of toxicity. In this work, we investigated the physiological and toxicological responses of silkworm to combined oral administration of CAM and VCM. Results showed that antibiotics promote the feeding behavior of silkworm and significantly reduce (P < 0.05) intestinal cultivable bacterial counts. Moreover, antibiotics decreased the antioxidant enzyme activities of superoxide dismutase, catalase, glutathione S-transferase, and thioredoxin reductase and caused oxidative damage to the silkworm intestine; the degree of damage was confirmed by histopathology analysis. The gene expression levels of antimicrobial peptides (attacin, lysozyme, and cecropins) were also perturbed by antibiotics. After antibiotic exposure, 16S rRNA metagenomic sequencing revealed increases in the relative abundance of Sphingobium, Burkholderia, Barnesiella, Bacteroides, Bradyrhizobium, Acinetobacter, Phenylobacterium, Plesiomonas, Escherichia/Shigella, and unclassified bacteria, as well as a reduction of Enterococcus. The metabolic and functional profiles of intestinal microbiota, particularly metabolic processes, such as energy, cofactors and vitamins, lipid, amino acid, and carbohydrate metabolisms, changed after antibiotic exposure. In conclusion, our findings reveal that antibiotics exert substantial effects on silkworm. The present study may promote the applications of silkworm as an interspecies model in the medical and pharmaceutical fields.
Collapse
Affiliation(s)
- Guannan Li
- State Key Laboratory of Silkworm Genome Biology, College of Biotechnology, Southwest University, Chongqing, 400716, China.
| | - Xuejuan Xia
- Food Science and Technology Programme, Department of Chemistry, National University of Singapore, Singapore, 117546, Singapore
| | - Shan Zhao
- State Key Laboratory of Silkworm Genome Biology, College of Biotechnology, Southwest University, Chongqing, 400716, China
| | - Min Shi
- State Key Laboratory of Silkworm Genome Biology, College of Biotechnology, Southwest University, Chongqing, 400716, China
| | - Fengdan Liu
- State Key Laboratory of Silkworm Genome Biology, College of Biotechnology, Southwest University, Chongqing, 400716, China
| | - Yong Zhu
- State Key Laboratory of Silkworm Genome Biology, College of Biotechnology, Southwest University, Chongqing, 400716, China.
| |
Collapse
|
154
|
Emberger J, Hitchcock MM, Markley JD. Diagnostic Stewardship Approaches to Clostridioides difficile Infection in the Era of Two-Step Testing: a Shifting Landscape. CURRENT TREATMENT OPTIONS IN INFECTIOUS DISEASES 2020. [DOI: 10.1007/s40506-020-00223-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
155
|
Liu H, Wang HH. Impact of Microbiota Transplant on Resistome of Gut Microbiota in Gnotobiotic Piglets and Human Subjects. Front Microbiol 2020; 11:932. [PMID: 32508773 PMCID: PMC7248251 DOI: 10.3389/fmicb.2020.00932] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/20/2020] [Indexed: 12/20/2022] Open
Abstract
Microbiota transplant is becoming a popular process to restore or initiate “healthy” gut microbiota and immunity. But, the potential risks of the related practices need to be carefully evaluated. This study retrospectively examined the resistomes of donated fecal microbiota for treating intestinal disorders, vaginal microbiota of pregnant women, and infant fecal microbiota from rural and urban communities, as well as the impact of transplants on the fecal resistome of human and animal recipients. Antibiotic resistance (AR) genes were found to be abundant in all donor microbiota. An overall surge of resistomes with higher prevalence and abundance of AR genes was observed in the feces of all transplanted gnotobiotic pigs as well as in the feces of infant subjects, compared to those in donor fecal and maternal vaginal microbiota. Surprisingly, transplants using rural Amish microbiota led to more instead of less AR genes in the fecal microbiota of gnotobiotic pigs than did transplants using urban microbiota. New AR gene subtypes undetected originally also appeared in gnotobiotic pigs, in Crohn’s Disease (CD) patients after transplant, and in feces of infant subjects. The data illustrated the key role of the host gastrointestinal tract system in amplifying the ever-increasing AR gene pool, even without antibiotic exposure. The data further suggest that the current approaches of microbiota transplant can introduce significant health risk factor(s) to the recipients, and newborn human and animal hosts with naïve gut microbiota were especially susceptible. Given the illustrated public health risks of microbiota transplant, minimizing massive and unnecessary damages to gut microbiota by oral antibiotics and other gut impacting drugs becomes important. Since eliminating risk factors including AR bacteria and opportunistic pathogens directly from donor microbiota is still difficult to achieve, developing microbial cocktails with defined organisms and functions has further become an urgent need, should microbiota transplantation become necessary.
Collapse
Affiliation(s)
- Hu Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China.,Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States
| | - Hua H Wang
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States.,Department of Microbiology, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
156
|
Rastogi YR, Saini AK, Thakur VK, Saini RV. New Insights into Molecular Links Between Microbiota and Gastrointestinal Cancers: A Literature Review. Int J Mol Sci 2020; 21:E3212. [PMID: 32370077 PMCID: PMC7246717 DOI: 10.3390/ijms21093212] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/20/2022] Open
Abstract
Despite decades of exhaustive research on cancer, questions about cancer initiation, development, recurrence, and metastasis have still not been completely answered. One of the reasons is the plethora of factors acting simultaneously in a tumour microenvironment, of which not all have garnered attention. One such factor that has long remained understudied and has only recently received due attention is the host microbiota. Our sheer-sized microbiota exists in a state of symbiosis with the body and exerts significant impact on our body's physiology, ranging from immune-system development and regulation to neurological and cognitive development. The presence of our microbiota is integral to our development, but a change in its composition (microbiota dysbiosis) can often lead to adverse effects, increasing the propensity of serious diseases like cancers. In the present review, we discuss environmental and genetic factors that cause changes in microbiota composition, disposing of the host towards cancer, and the molecular mechanisms (such as β-catenin signalling) and biochemical pathways (like the generation of oncogenic metabolites like N-nitrosamines and hydrogen sulphide) that the microbiota uses to initiate or accelerate cancers, with emphasis on gastrointestinal cancers. Moreover, we discuss how microbiota can adversely influence the success of colorectal-cancer chemotherapy, and its role in tumour metastasis. We also attempted to resolve conflicting results obtained for the butyrate effect on tumour suppression in the colon, often referred to as the 'butyrate paradox'. In addition, we suggest the development of microbiota-based biomarkers for early cancer diagnosis, and a few target molecules of which the inhibition can increase the overall chances of cancer cure.
Collapse
Affiliation(s)
- Yash Raj Rastogi
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh 173229, India;
| | - Adesh K. Saini
- Faculty of Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh 173229, India;
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Centre, Scotland’s Rural College (SRUC), Kings Buildings, Edinburgh, EH9 3JG, UK
| | - Reena V. Saini
- School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh 173229, India
| |
Collapse
|
157
|
Scher JU, Nayak RR, Ubeda C, Turnbaugh PJ, Abramson SB. Pharmacomicrobiomics in inflammatory arthritis: gut microbiome as modulator of therapeutic response. Nat Rev Rheumatol 2020; 16:282-292. [PMID: 32157196 PMCID: PMC11221369 DOI: 10.1038/s41584-020-0395-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2020] [Indexed: 02/07/2023]
Abstract
In the past three decades, extraordinary advances have been made in the understanding of the pathogenesis of, and treatment options for, inflammatory arthritides, including rheumatoid arthritis and spondyloarthritis. The use of methotrexate and subsequently biologic therapies (such as TNF inhibitors, among others) and oral small molecules have substantially improved clinical outcomes for many patients with inflammatory arthritis; for others, however, these agents do not substantially improve their symptoms. The emerging field of pharmacomicrobiomics, which investigates the effect of variations within the human gut microbiome on drugs, has already provided important insights into these therapeutics. Pharmacomicrobiomic studies have demonstrated that human gut microorganisms and their enzymatic products can affect the bioavailability, clinical efficacy and toxicity of a wide array of drugs through direct and indirect mechanisms. This discipline promises to facilitate the advent of microbiome-based precision medicine approaches in inflammatory arthritis, including strategies for predicting response to treatment and for modulating the microbiome to improve response to therapy or reduce drug toxicity.
Collapse
Affiliation(s)
- Jose U Scher
- Department of Medicine, Division of Rheumatology, New York University Langone Health, New York, NY, USA.
| | - Renuka R Nayak
- Rheumatology Division, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology & Immunology, University of California, San Francisco, CA, USA
| | - Carles Ubeda
- Centro Superior de Investigacion en Salud Publica - FISABIO, Valencia, Spain
- CIBER en Epidemiologia y Salud Publica, Madrid, Spain
| | - Peter J Turnbaugh
- Department of Microbiology & Immunology, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Steven B Abramson
- Department of Medicine, Division of Rheumatology, New York University Langone Health, New York, NY, USA
| |
Collapse
|
158
|
Antibacterial Monoclonal Antibodies Do Not Disrupt the Intestinal Microbiome or Its Function. Antimicrob Agents Chemother 2020; 64:AAC.02347-19. [PMID: 32152087 PMCID: PMC7179586 DOI: 10.1128/aac.02347-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/05/2020] [Indexed: 02/07/2023] Open
Abstract
Antibiotics revolutionized the treatment of infectious diseases; however, it is now clear that broad-spectrum antibiotics alter the composition and function of the host’s microbiome. The microbiome plays a key role in human health, and its perturbation is increasingly recognized as contributing to many human diseases. Widespread broad-spectrum antibiotic use has also resulted in the emergence of multidrug-resistant pathogens, spurring the development of pathogen-specific strategies such as monoclonal antibodies (MAbs) to combat bacterial infection. Antibiotics revolutionized the treatment of infectious diseases; however, it is now clear that broad-spectrum antibiotics alter the composition and function of the host’s microbiome. The microbiome plays a key role in human health, and its perturbation is increasingly recognized as contributing to many human diseases. Widespread broad-spectrum antibiotic use has also resulted in the emergence of multidrug-resistant pathogens, spurring the development of pathogen-specific strategies such as monoclonal antibodies (MAbs) to combat bacterial infection. Not only are pathogen-specific approaches not expected to induce resistance in nontargeted bacteria, but they are hypothesized to have minimal impact on the gut microbiome. Here, we compare the effects of antibiotics, pathogen-specific MAbs, and their controls (saline or control IgG [c-IgG]) on the gut microbiome of 7-week-old, female, C57BL/6 mice. The magnitude of change in taxonomic abundance, bacterial diversity, and bacterial metabolites, including short-chain fatty acids (SCFA) and bile acids in the fecal pellets from mice treated with pathogen-specific MAbs, was no different from that with animals treated with saline or an IgG control. Conversely, dramatic changes were observed in the relative abundance, as well as alpha and beta diversity, of the fecal microbiome and bacterial metabolites in the feces of all antibiotic-treated mice. Taken together, these results indicate that pathogen-specific MAbs do not alter the fecal microbiome like broad-spectrum antibiotics and may represent a safer, more-targeted approach to antibacterial therapy.
Collapse
|
159
|
Jia L, Cao M, Chen H, Zhang M, Dong X, Ren Z, Sun J, Pan LL. Butyrate Ameliorates Antibiotic-Driven Type 1 Diabetes in the Female Offspring of Nonobese Diabetic Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:3112-3120. [PMID: 32046486 DOI: 10.1021/acs.jafc.9b07701] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Maternal gut dysbiosis affects the development of the offspring immune system. Our previous study has indicated that microbial metabolite butyrate directly shapes pancreatic immune tolerance and dampens type 1 diabetes (T1D) progression. Therefore, maternal butyrate intervention may protect their offspring from maternal gut dysbiosis-accelerated T1D. To test this, pregnant nonobese diabetic (NOD) mice were treated with vancomycin in drinking water with or without a butyrate-supplemented diet during gestation and nursing (oral vancomycin is used to induce maternal gut dysbiosis). Three weeks after delivery, T1D-associated innate and adaptive immune cells were detected to investigate the effects of butyrate on the vancomycin-exacerbated pancreatic immune disorder in dams and pups. The results showed that butyrate inhibited maternal vancomycin-exacerbated secretion of proinflammation cytokines (interferon γ and interleukin-1β) and maternal vancomycin-exacerbated recruitment of interferon γ+ T cells (cytotoxic T lymphocytes 1 cells and T helper type 1 cells) in the pancreas of the female offspring, thus dampening T1D development. The protection may be due to butyrate inhibiting the activation of pancreatic dendritic cells (DCs). Our data thus demonstrate that maternal gut dysbiosis can exacerbate pancreatic-directed autoimmunity in the female offspring through T cell- and DC-associated mechanisms that are inhibited by butyrate.
Collapse
Affiliation(s)
- Lingling Jia
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, P. R. China
| | - Minkai Cao
- Department of Obstetrics, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, Jiangsu, P. R. China
| | - Hao Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, P. R. China
| | - Ming Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, P. R. China
| | - Xiaoliang Dong
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, P. R. China
| | - Zhengnan Ren
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, P. R. China
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, P. R. China
| | - Li-Long Pan
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, P. R. China
| |
Collapse
|
160
|
Gut Microbiota, Antibiotic Therapy and Antimicrobial Resistance: A Narrative Review. Microorganisms 2020; 8:microorganisms8020269. [PMID: 32079318 PMCID: PMC7074698 DOI: 10.3390/microorganisms8020269] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/08/2020] [Accepted: 02/08/2020] [Indexed: 12/13/2022] Open
Abstract
Antimicrobial resistance is a major concern. Epidemiological studies have demonstrated direct relationships between antibiotic consumption and emergence/dissemination of resistant strains. Within the last decade, authors confounded spectrum activity and ecological effects and did not take into account several other factors playing important roles, such as impact on anaerobic flora, biliary elimination and sub-inhibitory concentration. The ecological impact of antibiotics on the gut microbiota by direct or indirect mechanisms reflects the breaking of the resistance barrier to colonization. To limit the impact of antibiotic therapy on gut microbiota, consideration of the spectrum of activity and route of elimination must be integrated into the decision. Various strategies to prevent (antimicrobial stewardship, action on residual antibiotics at colonic level) or cure dysbiosis (prebiotic, probiotic and fecal microbiota transplantation) have been introduced or are currently being developed.
Collapse
|
161
|
Wu KS, Syue LS, Cheng A, Yen TY, Chen HM, Chiu YH, Hsu YL, Chiu CH, Su TY, Tsai WL, Chen WY, Huang CH, Hung HM, Huang LJ, Kuo HJ, Lin PC, Yang CH, Hong PL, Lee SSJ, Chen YS, Liu YC, Huang LM. Recommendations and guidelines for the treatment of Clostridioides difficile infection in Taiwan. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2020; 53:191-208. [PMID: 32169531 DOI: 10.1016/j.jmii.2020.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/19/2020] [Accepted: 02/01/2020] [Indexed: 12/13/2022]
Abstract
Clostridioides difficile infection (CDI) is a major enteric disease associated with antibiotic use and a leading cause of hospital-acquired infections worldwide. This is the first guideline for treatment of CDI in Taiwan, aiming to optimize medical care for patients with CDI. The target audience of this document includes all healthcare personnel who are involved in the medical care of patients with CDI. The 2018 Guidelines Recommendations for Evidence-based Antimicrobial agents use in Taiwan (GREAT) working group was formed, comprising of infectious disease specialists from 13 medical centers in Taiwan, to review the evidence and draft recommendations using the grading of recommendations assessment, development, and evaluation (GRADE) methodology. A nationwide expert panel reviewed the recommendations during a consensus meeting in March 2019. The recommendation is endorsed by the Infectious Diseases Society of Taiwan (IDST). This guideline describes the epidemiology and risk factors of CDI, and provides recommendations for treatment of CDI in both adults and children. Recommendations for treatment of the first episode of CDI, first recurrence, second and subsequent recurrences of CDI, severe CDI, fulminant CDI, and pediatric CDI are provided.
Collapse
Affiliation(s)
- Kuan-Sheng Wu
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ling-Shan Syue
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Aristine Cheng
- Division of Infectious Diseases, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ting-Yu Yen
- Departments of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsien-Meng Chen
- Division of Infectious Disease, Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Yu-Hsin Chiu
- Division of Infectious Diseases, Chi-Mei Medical Center, Tainan, Taiwan
| | - Yu-Lung Hsu
- Division of Pediatric Infectious Diseases, China Medical University Children's Hospital, China Medical University, Taichung, Taiwan
| | - Chun-Hsiang Chiu
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ting-Yi Su
- Division of Infectious Diseases, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Wan-Lin Tsai
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Wei-Yu Chen
- Division of General Medicine, Infectious Disease, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Chung-Hao Huang
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Huei-Min Hung
- Department of Pediatrics, Chang Gung Children's Hospital, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ling-Ju Huang
- Division of General Medicine, Infectious Diseases, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hong-Jie Kuo
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Pei-Chin Lin
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Department of Pharmacy, School of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Hsiang Yang
- Department of Pharmacy, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Pi-Lien Hong
- Department of Pharmacy, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Susan Shin-Jung Lee
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| | - Yao-Shen Chen
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yung-Ching Liu
- Division of Infectious Diseases, Taipei Medical University Shuang Ho Hospital, Taipei, Taiwan
| | - Li-Ming Huang
- Departments of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
162
|
Jarmo O, Veli-Jukka A, Eero M. Treatment of Clostridioides (Clostridium) difficile infection. Ann Med 2020; 52:12-20. [PMID: 31801387 PMCID: PMC7877971 DOI: 10.1080/07853890.2019.1701703] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/29/2019] [Accepted: 11/24/2019] [Indexed: 02/06/2023] Open
Abstract
Clostridioides (formerly: Clostridium) difficile infection (CDI) is a major cause of diarrhoea for inpatients as well as outpatients. Usually, CDI is healthcare-associated but the number of community-acquired infections is increasing. CDI is generally associated with changes in the normal intestinal microbiota caused by administration of antibiotics. Elderly and immunocompromised patients are at greater risk for CDI and CDI recurrence. Recently, the treatment options of CDI have undergone major changes: current recommendations speak against using metronidazole for primary CDI, fidaxomicin and bezlotoxumab have been added to the treatment armamentarium and microbial replacement therapies have emerged. Several other therapies are undergoing clinical trials. In this article, we review current treatment guidelines, present the most recent data on the options to treat CDI and glance towards future developments.KEY MESSAGESThe cornerstones for the treatment of CDI are vancomycin and fidaxomicin. Metronidazole should be used only in mild-to-moderate disease in younger patients who have no or only few risk factors for recurrence.In recurrent CDI, bezlotoxumab infusion (a monoclonal antibody against C. difficile toxin B) may be considered as an adjunctive therapeutic strategy in addition to the standard care provided to patients with several risk factors for recurrence.Faecal microbiota transplantation (FMT) should be offered to patients with frequently recurring CDI.
Collapse
Affiliation(s)
- Oksi Jarmo
- Department of Infectious Diseases, Turku University Hospital, University of Turku, Turku, Finland
| | - Anttila Veli-Jukka
- Department of Infectious Diseases, Helsinki University Central Hospital, Helsinki, Finland
| | - Mattila Eero
- Department of Infectious Diseases, Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
163
|
Reply to: Caution is warranted in using cephamycin antibiotics against recurrent Clostridioides difficile infection. Nat Microbiol 2020; 5:237-238. [DOI: 10.1038/s41564-019-0662-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 12/15/2019] [Indexed: 11/08/2022]
|
164
|
Rauseo AM, Olsen MA, Reske KA, Dubberke ER. Strategies to prevent adverse outcomes following Clostridioides difficile infection in the elderly. Expert Rev Anti Infect Ther 2020; 18:203-217. [PMID: 31976779 DOI: 10.1080/14787210.2020.1717950] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction: Clostridioides difficile remains the most common cause of healthcare-associated infections in the US, and it disproportionately affects the elderly. Older patients are more susceptible and have a greater risk of adverse outcomes from C. difficile infection (CDI), despite advances in treatment and prevention.Areas covered: The epidemiology and pathogenesis of CDI, as well as risk factors in the aging host, will be reviewed. The importance of antimicrobial stewardship and infection prevention in order to avoid acquisition and transmission will be discussed, as well as strategies to prevent adverse outcomes and recurrent CDI, through optimization of CDI treatment s,election.Expert opinion: Appropriate CDI-prevention strategies to avoid adverse outcomes in this susceptible population involve antimicrobial stewardship and methods to prevent C. difficile transmission in healthcare settings. Management strategies to prevent adverse outcomes include initiation of supportive therapy and proper selection of CDI specific treatments. Many patients may also benefit from adjunctive therapies or additional procedures.
Collapse
Affiliation(s)
- Adriana M Rauseo
- Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, USA
| | - Margaret A Olsen
- Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, USA
| | - Kimberly A Reske
- Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, USA
| | - Erik R Dubberke
- Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
165
|
High Heterogeneity of Multidrug-Resistant Enterobacteriaceae Fecal Levels in Hospitalized Patients Is Partially Driven by Intravenous β-Lactams. Antimicrob Agents Chemother 2020; 64:AAC.01415-19. [PMID: 31767720 DOI: 10.1128/aac.01415-19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/12/2019] [Indexed: 11/20/2022] Open
Abstract
Multidrug-resistant Enterobacteriaceae (MRE) colonize the intestine asymptomatically from where they can breach into the bloodstream and cause life-threatening infections, especially in heavily colonized patients. Despite the clinical relevance of MRE colonization levels, we know little about how they vary in hospitalized patients and the clinical factors that determine those levels. Here, we conducted one of the largest studies of MRE fecal levels by tracking longitudinally 133 acute leukemia patients and monitoring their MRE levels over time through extensive culturing. MRE were defined as Enterobacteriaceae species that acquired nonsusceptibility to ≥1 agent in ≥3 antimicrobial categories. In addition, due to the selective media used, the MRE had to be resistant to third-generation cephalosporins. MRE were detected in 60% of the patients, but their fecal levels varied considerably among patients and within the same patient (>6 and 4 orders of magnitude, respectively). Multivariate analysis of clinical metadata revealed an impact of intravenous beta-lactams (i.e., meropenem and piperacillin-tazobactam), which significantly diminished the fecal MRE levels in hospitalized patients. Consistent with a direct action of beta-lactams, we found an effect only when the patient was colonized with strains sensitive to the administered beta-lactam (P < 0.001) but not with nonsusceptible strains. We report previously unobserved inter- and intraindividual heterogeneity in MRE fecal levels, suggesting that quantitative surveillance is more informative than qualitative surveillance of hospitalized patients. In addition, our study highlights the relevance of incorporating antibiotic treatment and susceptibility data of gut-colonizing pathogens for future clinical studies and in clinical decision-making.
Collapse
|
166
|
Zhu Y, Liu K, Zhang J, Liu X, Yang L, Wei R, Wang S, Zhang D, Xie S, Tao F. Antibiotic body burden of elderly Chinese population and health risk assessment: A human biomonitoring-based study. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 256:113311. [PMID: 31813705 DOI: 10.1016/j.envpol.2019.113311] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 09/24/2019] [Accepted: 09/25/2019] [Indexed: 06/10/2023]
Abstract
Recently, the widespread use of antibiotic has raised concerns about the potential health risks associated with their microbiological effect. In the present study, we investigated 990 elderly individuals (age ≥ 60 years) from the Cohort of Elderly Health and Environment Controllable Factors in West Anhui, China. A total of 45 representative antibiotics and two antibiotic metabolites were monitored in urine samples through liquid chromatography electrospray tandem mass spectrometry. The results revealed that 34 antibiotics were detected in 93.0% of all urine samples and the detection frequencies of each antibiotic varied between 0.2% and 35.5%. The overall detection frequencies of seven human antibiotics (HAs), 10 veterinary antibiotics (VAs), three antibiotics preferred as HAs (PHAs), and 14 preferred as VAs (PVAs) in urines were 27.4%, 62.9%, 30.9% and 72.7%, respectively. Notably, the samples with concentrations of six PVAs (sulfamethoxazole, trimethoprim, oxytetracycline, danofloxacin, norfloxacin and lincomycin) above 5000 ng/mL accounted for 1.7% of all urine samples. Additionally, in 62.7% of urine samples, the total antibiotic concentration was in the range of the limits of detection to 20.0 ng/mL. Furthermore, the elderly individuals with the sum of estimated daily intakes of VAs and PVAs more than 1 μg/kg/day accounted for 15.2% of all participants, and a health risk related to change in gut microbiota under antibiotic stimulation was expected in 6.7% of the elderly individuals. Especially, ciprofloxacin was the foremost contributor to the health risk, and its hazard quotient value was more than one in 3.5% of all subjects. Taken together, the elderly Chinese people were extensively exposed to VAs, and some elderly individuals may have a health risk associated with dysbiosis of the gut microbiota.
Collapse
Affiliation(s)
- Yitian Zhu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Kaiyong Liu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jingjing Zhang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Xinji Liu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Linsheng Yang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Rong Wei
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Sufang Wang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Dongmei Zhang
- School of Health Management, Anhui Medical University, Hefei, 230032, China
| | - Shaoyu Xie
- Lu'an Center of Disease Control and Prevention, Lu'an, Anhui, 237000, PR China
| | - Fangbiao Tao
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
167
|
McGovern AS, Hamlin AS, Winter G. A review of the antimicrobial side of antidepressants and its putative implications on the gut microbiome. Aust N Z J Psychiatry 2019; 53:1151-1166. [PMID: 31558039 DOI: 10.1177/0004867419877954] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Serotonin reuptake inhibitors are the predominant treatment for major depressive disorder. In recent years, the diversity of the gut microbiota has emerged to play a significant role in the occurrence of major depressive disorder and other mood and anxiety disorders. Importantly, the role of the gut microbiota in the treatment of such disorders remains to be elucidated. Here, we provide a review of the literature regarding the effects of physiologically relevant concentrations of serotonin reuptake inhibitors on the gut microbiota and the implications this might have on their efficacy in the treatment of mood disorders. METHODS First, an estimation of gut serotonin reuptake inhibitor concentrations was computed based on pharmacokinetic and gastrointestinal transit properties of serotonin reuptake inhibitors. Literature regarding the in vivo and in vitro antimicrobial properties of serotonin reuptake inhibitors was gathered, and the estimated gut concentrations were examined in the context of these data. Computer-based investigation revealed putative mechanisms for the antimicrobial effects of serotonin reuptake inhibitors. RESULTS In vivo evidence using animal models shows an antimicrobial effect of serotonin reuptake inhibitors on the gut microbiota. Examination of the estimated physiological concentrations of serotonin reuptake inhibitors in the gastrointestinal tract collected from in vitro studies suggests that the microbial community of both the small intestine and the colon are exposed to serotonin reuptake inhibitors for at least 4 hours per day at concentrations that are likely to exert an antimicrobial effect. The potential mechanisms of the effect of serotonin reuptake inhibitors on the gut microbiota were postulated to include inhibition of efflux pumps and/or amino acid transporters. CONCLUSION This review raises important issues regarding the role that gut microbiota play in the treatment of mood-related behaviours, which holds substantial potential clinical outcomes for patients suffering from major depressive disorder and other mood-related disorders.
Collapse
Affiliation(s)
- Abigail S McGovern
- School of Science and Technology, University of New England, Armidale, NSW, Australia
| | - Adam S Hamlin
- School of Science and Technology, University of New England, Armidale, NSW, Australia
| | - Gal Winter
- School of Science and Technology, University of New England, Armidale, NSW, Australia
| |
Collapse
|
168
|
Wilcox MH, Cornely OA, Guery B, Longshaw C, Georgopali A, Karas A, Kazeem G, Palacios-Fabrega JA, Vehreschild MJGT. Microbiological Characterization and Clinical Outcomes After Extended-Pulsed Fidaxomicin Treatment for Clostridioides difficile Infection in the EXTEND Study. Open Forum Infect Dis 2019; 6:ofz436. [PMID: 31723569 PMCID: PMC6834086 DOI: 10.1093/ofid/ofz436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 11/05/2019] [Indexed: 11/17/2022] Open
Abstract
Background Clostridioides (Clostridium) difficile infection (CDI) is diagnosed using clinical signs and symptoms plus positive laboratory tests. Recurrence of CDI after treatment is common, and coinfection with other enteric pathogens may influence clinical outcomes. Methods We aimed to assess rates of C difficile positivity, by enzyme-linked immunosorbent assay (ELISA) toxin A/B and BioFire FilmArray, and the effect of enteric coinfection on clinical outcomes, using samples from the EXTEND study of extended-pulsed fidaxomicin (EPFX) versus standard vancomycin. Results All 356 randomized and treated patients tested positive for C difficile toxin A/B by local tests; a majority (225 of 356, 63.2%) also tested positive by both ELISA and BioFire. Most stool samples taken at screening tested positive for C difficile only using BioFire (EPFX: 112 of 165, 69.7%; vancomycin: 118 of 162, 72.8%). Of the 5 patients who failed treatment and had stool samples available, all (1) had tested negative for C difficile by BioFire at screening and (2) were negative by ELISA at time of treatment failure. When analyzed by BioFire results at screening, rates of sustained clinical cure at 30 days after end of treatment were numerically higher with EPFX than with vancomycin for almost all patients, except for those who tested negative for C difficile but positive for another pathogen. However, these outcome differences by presence of coinfection did not reach statistical significance. Whole-genome sequencing analysis determined that 20 of 26 paired samples from patients with recurrence were reinfections with the same C difficile strain. Conclusions Testing for presence of copathogens in clinical trials of antibiotics could help to explain clinical failures.
Collapse
Affiliation(s)
- Mark H Wilcox
- Department of Microbiology, Leeds Teaching Hospitals and University of Leeds, Leeds, United Kingdom.,Healthcare Associated Infections Research Group, Section of Molecular Gastroenterology, Leeds Institute for Biomedical and Clinical Sciences, University of Leeds, United Kingdom
| | - Oliver A Cornely
- Clinical Trials Centre Cologne, ZKS Köln, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany.,Department of Internal Medicine, University Hospital of Cologne and German Centre for Infection Research, Partner Site Bonn-Cologne, Cologne, Germany
| | - Benoit Guery
- Infectious Diseases Service, Department of Medicine, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Chris Longshaw
- Astellas Pharma Europe Ltd., Chertsey, United Kingdom.,Department of Microbiology, Leeds Teaching Hospitals and University of Leeds, Leeds, United Kingdom
| | | | | | - Gbenga Kazeem
- Astellas Pharma Europe Ltd., Chertsey, United Kingdom
| | | | - Maria J G T Vehreschild
- Department of Internal Medicine, University Hospital of Cologne and German Centre for Infection Research, Partner Site Bonn-Cologne, Cologne, Germany.,Medical Clinic II, University Hospital Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
169
|
Adams JB, Borody TJ, Kang DW, Khoruts A, Krajmalnik-Brown R, Sadowsky MJ. Microbiota transplant therapy and autism: lessons for the clinic. Expert Rev Gastroenterol Hepatol 2019; 13:1033-1037. [PMID: 31665947 DOI: 10.1080/17474124.2019.1687293] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: The purpose of this review is to discuss Microbiota Transplant Therapy (MTT), a type of intensive intestinal microbiota transplantation (IMT), for people with autism spectrum disorders (ASD) and chronic gastrointestinal disorders (constipation and/or diarrhea).Areas covered: This paper briefly reviews IMT, gastrointestinal symptoms and gastrointestinal bacteria in children with ASD, and results and lessons learned from intensive MTT for autism.Expert opinion: An open-label study and a two-year follow-up suggest that MTT is relatively safe and effective in significantly reducing gastrointestinal disorders and autism symptoms, changing the gut microbiome structure, and increasing gut microbial diversity. Further research with larger, randomized, double-blind, placebo-controlled studies is warranted.
Collapse
Affiliation(s)
- James B Adams
- School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, Arizona, USA
| | | | - Dae-Wook Kang
- Department of Civil & Environmental Engineering, The University of Toledo, Toledo, Ohio, USA
| | - Alexander Khoruts
- Department of Medicine, Division of Gastroenterology; Center for Immunology, BioTechnology Institute, University of Minnesota, Minnepolis, Minnesota, USA
| | - Rosa Krajmalnik-Brown
- Swette Center for Environmental Biotechnology, Arizona State University, Tempe, Arizona, USA
| | - Michael J Sadowsky
- BioTechnology Institute, and Departments of Soil, Water & Climate, and Plant& Microbial Biology, University of Minnesota, St. Paul, Minnesota, USA
| |
Collapse
|
170
|
Avni T, Babitch T, Ben-Zvi H, Hijazi R, Ayada G, Atamna A, Bishara J. Clostridioides difficile infection in immunocompromised hospitalized patients is associated with a high recurrence rate. Int J Infect Dis 2019; 90:237-242. [PMID: 31672656 DOI: 10.1016/j.ijid.2019.10.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/20/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Clostridioides difficile infection (CDI) may pose a serious threat to immunocompromised patients (IMC). Herein, we evaluated the clinical outcomes of IMC patients with CDI. METHODS All consecutive hospitalized patients between January 1, 2013 and December 31, 2018 with laboratory confirmed CDI were included in the study. Subjects were divided into two groups: IMC patients and controls. Primary outcome was the recurrence rate of CDI (rCDI) at 30 and 90 days after the first CDI episode. Secondary outcomes included 30 and 90 day all-cause mortality, length of hospital stay (LOS) and readmission rates. A multivariate analysis adjusted other risk factors for recurrence. An analysis of IMC patient subgroups (based on type of IMC conditions) was also performed. Results are reported as odds ratios (OR) with a 95% confidence interval (95% CI). RESULTS A total of 573 patients were included, amongst them 149 IMC patients (36 solid organ transplants, 38 undergoing chemotherapy, 62 haematological conditions, 13 receiving high dose prednisone) and 424 controls. IMC patients were younger, independent and exhibited less significant comorbidities. On multivariable analysis, the rate of rCDI was significantly higher in IMC patients (OR 2.7, 95% CI 1.6-5). rCDI was also associated with vancomycin therapy, haemodialysis and previous hospitalizations. Mortality, LOS, CDI complications and rehospitalization rates were similar in both. CONCLUSIONS IMC patients with CDI have an increased risk of 90 days rCDI. Vancomycin treatment for CDI endangers recurrence in IMC patients. Further research should explore other therapies for IMC patients with CDI with alternative agents such as Fidaxomicin and Bezlotoxumab.
Collapse
Affiliation(s)
- Tomer Avni
- Infectious Disease Unit, Rabin Medical Center, Beilinson Hospital, Petah-Tikva, Israel; Internal Medicine Department E, Rabin Medical Center, Beilinson Hospital, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Tanya Babitch
- Internal Medicine Department E, Rabin Medical Center, Beilinson Hospital, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Haim Ben-Zvi
- Clinical Microbiology Laboratory, Rabin Medical Center, Beilinson Hospital, Petah-Tikva, Israel
| | - Rabab Hijazi
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Gida Ayada
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Alaa Atamna
- Infectious Disease Unit, Rabin Medical Center, Beilinson Hospital, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel.
| | - Jihad Bishara
- Infectious Disease Unit, Rabin Medical Center, Beilinson Hospital, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| |
Collapse
|
171
|
Baggs J, Jernigan JA, Halpin AL, Epstein L, Hatfield KM, McDonald LC. Risk of Subsequent Sepsis Within 90 Days After a Hospital Stay by Type of Antibiotic Exposure. Clin Infect Dis 2019; 66:1004-1012. [PMID: 29136126 DOI: 10.1093/cid/cix947] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 11/01/2017] [Indexed: 12/13/2022] Open
Abstract
Background We examined the risk of sepsis within 90 days after discharge from a previous hospital stay by type of antibiotic received during the previous stay. Methods We retrospectively identified a cohort of hospitalized patients from the Truven Health MarketScan Hospital Drug Database. We examined the association between the use of certain antibiotics during the initial hospital stay, determined a priori, and the risk of postdischarge sepsis controlling for potential confounding factors in a multivariable logistic regression model. Our primary exposure was receipt of antibiotics more strongly associated with clinically important microbiome disruption. Our primary outcome was a hospital stay within 90 days of the index stay that included an International Classification of Diseases, Ninth Revision, Clinical Modification (ICD-9-CM) discharge diagnosis of severe sepsis (ICD-9-CM code 995.92) or septic shock (785.52). Results Among 516 hospitals, we randomly selected a single stay for eligible patients. In 0.17% of these patients, severe sepsis/septic shock developed within 90 days after discharge. The risk of sepsis associated with exposure to our high-risk antibiotics was 65% higher than in those without antibiotic exposure. Conclusions Our study identified an increased risk of sepsis within 90 days of discharge among patients with exposure to high-risk antibiotics or increased quantities of antibiotics during hospitalization. Given that a significant proportion of inpatient antimicrobial use may be unnecessary, this study builds on previous evidence suggesting that increased stewardship efforts in hospitals may not only prevent antimicrobial resistance, Clostridium difficile infection, and other adverse effects, but may also reduce unwanted outcomes potentially related to disruption of the microbiota, including sepsis.
Collapse
Affiliation(s)
- James Baggs
- Division of Healthcare Quality Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - John A Jernigan
- Division of Healthcare Quality Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Alison Laufer Halpin
- Division of Healthcare Quality Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Lauren Epstein
- Division of Healthcare Quality Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Kelly M Hatfield
- Division of Healthcare Quality Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - L Clifford McDonald
- Division of Healthcare Quality Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
172
|
Pezo RC, Wong M, Martin A. Impact of the gut microbiota on immune checkpoint inhibitor-associated toxicities. Therap Adv Gastroenterol 2019; 12:1756284819870911. [PMID: 31555343 PMCID: PMC6747860 DOI: 10.1177/1756284819870911] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 07/17/2019] [Indexed: 02/04/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the treatment of patients with advanced cancers. However, the majority of patients do not respond or develop early progressive disease. A substantial number also develop immune-mediated toxicities that may lead to early treatment discontinuation. Gastrointestinal toxicities in the form of diarrhea and colitis are common and may resemble that observed in patients with inflammatory bowel disease (IBD). Alterations in the gut microbiota are thought to play an important role in mediating the intestinal inflammation that is associated with immune-mediated colitis. In this review, the authors' objective is to provide an overview of the gastrointestinal and hepatic toxicities that can be seen with ICIs and discuss the interactions between gut microbiota and the immune response. The authors also highlight the potential role for fecal microbial transfer (FMT) as an approach to improve therapeutic efficacy and decrease toxicity.
Collapse
Affiliation(s)
| | - Matthew Wong
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Alberto Martin
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
173
|
Ingham AC, Kielsen K, Cilieborg MS, Lund O, Holmes S, Aarestrup FM, Müller KG, Pamp SJ. Specific gut microbiome members are associated with distinct immune markers in pediatric allogeneic hematopoietic stem cell transplantation. MICROBIOME 2019; 7:131. [PMID: 31519210 PMCID: PMC6744702 DOI: 10.1186/s40168-019-0745-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/29/2019] [Indexed: 05/11/2023]
Abstract
BACKGROUND Increasing evidence reveals the importance of the microbiome in health and disease and inseparable host-microbial dependencies. Host-microbe interactions are highly relevant in patients receiving allogeneic hematopoietic stem cell transplantation (HSCT), i.e., a replacement of the cellular components of the patients' immune system with that of a foreign donor. HSCT is employed as curative immunotherapy for a number of non-malignant and malignant hematologic conditions, including cancers such as acute lymphoblastic leukemia. The procedure can be accompanied by severe side effects such as infections, acute graft-versus-host disease (aGvHD), and death. Here, we performed a longitudinal analysis of immunological markers, immune reconstitution and gut microbiota composition in relation to clinical outcomes in children undergoing HSCT. Such an analysis could reveal biomarkers, e.g., at the time point prior to HSCT, that in the future could be used to predict which patients are of high risk in relation to side effects and clinical outcomes and guide treatment strategies accordingly. RESULTS In two multivariate analyses (sparse partial least squares regression and canonical correspondence analysis), we identified three consistent clusters: (1) high concentrations of the antimicrobial peptide human beta-defensin 2 (hBD2) prior to the transplantation in patients with high abundances of Lactobacillaceae, who later developed moderate or severe aGvHD and exhibited high mortality. (2) Rapid reconstitution of NK and B cells in patients with high abundances of obligate anaerobes such as Ruminococcaceae, who developed no or mild aGvHD and exhibited low mortality. (3) High inflammation, indicated by high levels of C-reactive protein, in patients with high abundances of facultative anaerobic bacteria such as Enterobacteriaceae. Furthermore, we observed that antibiotic treatment influenced the bacterial community state. CONCLUSIONS We identify multivariate associations between specific microbial taxa, host immune markers, immune cell reconstitution, and clinical outcomes in relation to HSCT. Our findings encourage further investigations into establishing longitudinal surveillance of the intestinal microbiome and relevant immune markers, such as hBD2, in HSCT patients. Profiling of the microbiome may prove useful as a prognostic tool that could help identify patients at risk of poor immune reconstitution and adverse outcomes, such as aGvHD and death, upon HSCT, providing actionable information in guiding precision medicine.
Collapse
Affiliation(s)
- Anna Cäcilia Ingham
- Research Group for Genomic Epidemiology, Technical University of Denmark, Kongens Lyngby, Denmark
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Katrine Kielsen
- Institute for Inflammation Research, Department of Rheumatology and Spine Disease, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Malene Skovsted Cilieborg
- Comparative Pediatrics and Nutrition, Department of Clinical Veterinary and Animal Science, University of Copenhagen, Frederiksberg, Denmark
| | - Ole Lund
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Susan Holmes
- Department of Statistics, Stanford University, Stanford, USA
| | - Frank M Aarestrup
- Research Group for Genomic Epidemiology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Klaus Gottlob Müller
- Institute for Inflammation Research, Department of Rheumatology and Spine Disease, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Sünje Johanna Pamp
- Research Group for Genomic Epidemiology, Technical University of Denmark, Kongens Lyngby, Denmark.
| |
Collapse
|
174
|
Abstract
Many studies have shown that the urinary tract harbours its own microbial community known as the urinary microbiota, which have been implicated in urinary tract disorders. This observation contradicts the long-held notion that urine is a sterile biofluid in the absence of acute infection of the urinary tract. In light of this new discovery, many basic questions that are crucial for understanding the role of the urinary microbiota in human health and disease remain unanswered. Given that the urinary microbiota is an emerging area of study, optimized techniques and protocols to identify microorganisms in the urinary tract are still being established. However, the low microbial biomass and close proximity to higher microbial biomass environments (for example, the vagina) present distinct methodological challenges for microbial community profiling of the urinary microbiota. A clear understanding of the unique technical considerations for obtaining and analysing low microbial biomass samples, as well the influence of key elements of experimental design and computational analysis on downstream interpretation, will improve our ability to interpret and compare results across methods and studies and is relevant for studies profiling the urinary microbiota and other sites of low microbial abundance.
Collapse
|
175
|
Abstract
Oral rotavirus vaccines have consistently underperformed in low-income countries. In this issue of Cell Host & Microbe,Harris et al. (2018b) explore whether vaccine response can be enhanced via antibiotic-mediated modification of the bacterial microbiota.
Collapse
Affiliation(s)
- Edward P K Parker
- Department of Infectious Disease Epidemiology, St Mary's Campus, Imperial College London, London W2 1PG, UK.
| | - Nicholas C Grassly
- Department of Infectious Disease Epidemiology, St Mary's Campus, Imperial College London, London W2 1PG, UK
| |
Collapse
|
176
|
Chong PP, Koh AY. The gut microbiota in transplant patients. Blood Rev 2019; 39:100614. [PMID: 31492463 DOI: 10.1016/j.blre.2019.100614] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/22/2019] [Accepted: 08/27/2019] [Indexed: 02/09/2023]
Abstract
Solid organ transplant (SOT) and hematopoietic stem cell transplant (HSCT) recipients are at increased risk for developing infections due to underlying immunosuppression. Antibiotic use, and in HSCT recipients, the use of preparative regimens prior to transplantation can deplete gut commensal bacteria, resulting in intestinal dysbiosis. Emerging evidence in transplant patients, particularly HSCT, suggest that disturbances in gut microbiota populations are associated with a number of adverse outcomes. Here, we review the outcomes of HSCT and SOT recipients with gut microbiota imbalance or dysbiosis, explore the nascent field of gut microbiome therapeutic approaches including fecal microbiota transplantation and the use of precision probiotics in HSCT and SOT recipients.
Collapse
Affiliation(s)
- Pearlie P Chong
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Andrew Y Koh
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Harold C. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
177
|
Tan X, Johnson S. Fecal microbiota transplantation (FMT) for C. difficile infection, just say 'No'. Anaerobe 2019; 60:102092. [PMID: 31472233 DOI: 10.1016/j.anaerobe.2019.102092] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/14/2019] [Accepted: 08/22/2019] [Indexed: 12/11/2022]
Abstract
Despite lack of regulatory approval, fecal microbiota transplantation (FMT) is widely performed to manage C. difficile infection (CDI), particularly recurrent CDI. Herein, we critically review the available randomized controlled trials of FMT and address the following questions: Is FMT better than drug management of recurrent CDI?; Is FMT treatment per se or adjunctive treatment to antibiotics for CDI?; and, Is FMT safe? Finally, we elaborate non-FMT options for the management of recurrent CDI. Although promising, FMT should be reserved for patients who have failed appropriate antibiotic management of recurrent CDI.
Collapse
Affiliation(s)
- Xing Tan
- University of Illinois at Chicago, Chicago, IL, USA
| | - Stuart Johnson
- Edward Hines, Jr. VA Hospital, Hines, IL, USA; Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA.
| |
Collapse
|
178
|
Hasan N, Yang H. Factors affecting the composition of the gut microbiota, and its modulation. PeerJ 2019; 7:e7502. [PMID: 31440436 PMCID: PMC6699480 DOI: 10.7717/peerj.7502] [Citation(s) in RCA: 432] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/17/2019] [Indexed: 12/13/2022] Open
Abstract
Gut microbiota have important functions in the body, and imbalances in the composition and diversity of those microbiota can cause several diseases. The host fosters favorable microbiota by releasing specific factors, such as microRNAs, and nonspecific factors, such as antimicrobial peptides, mucus and immunoglobulin A that encourage the growth of specific types of bacteria and inhibit the growth of others. Diet, antibiotics, and age can change gut microbiota, and many studies have shown the relationship between disorders of the microbiota and several diseases and reported some ways to modulate that balance. In this review, we highlight how the host shapes its gut microbiota via specific and nonspecific factors, how environmental and nutritional factors affect it, and how to modulate it using prebiotics, probiotics, and fecal microbiota transplantation.
Collapse
Affiliation(s)
- Nihal Hasan
- Department of Microbiology, Northeast Forestry University, Harbin, Heilongjiang, China.,Faculty of Health Science, Al-Baath University, Homs, Syria
| | - Hongyi Yang
- Department of Microbiology, Northeast Forestry University, Harbin, Heilongjiang, China
| |
Collapse
|
179
|
Taylor MR, Flannigan KL, Rahim H, Mohamud A, Lewis IA, Hirota SA, Greenway SC. Vancomycin relieves mycophenolate mofetil-induced gastrointestinal toxicity by eliminating gut bacterial β-glucuronidase activity. SCIENCE ADVANCES 2019; 5:eaax2358. [PMID: 31457102 PMCID: PMC6685722 DOI: 10.1126/sciadv.aax2358] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/28/2019] [Indexed: 05/08/2023]
Abstract
Mycophenolate mofetil (MMF) is commonly prescribed and has proven advantages over other immunosuppressive drugs. However, frequent gastrointestinal side effects through an unknown mechanism limit its use. We have found that consumption of MMF alters the composition of the gut microbiota, selecting for bacteria expressing the enzyme β-glucuronidase (GUS) and leading to an up-regulation of GUS activity in the gut of mice and symptomatic humans. In the mouse, vancomycin eliminated GUS-expressing bacteria and prevented MMF-induced weight loss and colonic inflammation. Our work provides a mechanism for the toxicity associated with MMF and a future direction for the development of therapeutics.
Collapse
Affiliation(s)
- Michael R. Taylor
- Departments of Pediatrics and Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kyle L. Flannigan
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Hannah Rahim
- Departments of Pediatrics and Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Amina Mohamud
- Departments of Pediatrics and Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ian A. Lewis
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Simon A. Hirota
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Steven C. Greenway
- Departments of Pediatrics and Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
180
|
Major G, Bradshaw L, Boota N, Sprange K, Diggle M, Montgomery A, Jawhari A, Spiller RC. Follow-on RifAximin for the Prevention of recurrence following standard treatment of Infection with Clostridium Difficile (RAPID): a randomised placebo controlled trial. Gut 2019; 68:1224-1231. [PMID: 30254135 PMCID: PMC6582824 DOI: 10.1136/gutjnl-2018-316794] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Clostridium difficile infection (CDI) recurs after initial treatment in approximately one in four patients. A single-centre pilot study suggested that this could be reduced using 'follow-on' rifaximin treatment. We aimed to assess the efficacy of rifaximin treatment in preventing recurrence. METHODS A multisite, parallel group, randomised, placebo controlled trial recruiting patients aged ≥18 years immediately after resolution of CDI through treatment with metronidazole or vancomycin. Participants received either rifaximin 400 mg three times a day for 2 weeks, reduced to 200 mg three times a day for a further 2 weeks or identical placebo. The primary endpoint was recurrence of CDI within 12 weeks of trial entry. RESULTS Between December 2012 and March 2016, 151 participants were randomised to either rifaximin or placebo. Primary outcome data were available on 130. Mean age was 71.9 years (SD 15.3). Recurrence within 12 weeks was 29.5% (18/61) among participants allocated to placebo compared with 15.9% (11/69) among those allocated to rifaximin, a difference between groups of 13.7% (95% CI -28.1% to 0.7%, p=0.06). The risk ratio was 0.54 (95% CI 0.28 to 1.05, p=0.07). During 6-month safety follow-up, nine participants died in each group (12%). Adverse event rates were similar between groups. CONCLUSION While 'follow-on' rifaximin after CDI appeared to halve recurrence rate, we failed to reach our recruitment target in this group of frail elderly patients, so the estimated effect of rifaximin lacks precision. A meta-analysis including a previous trial suggests that rifaximin may be effective; however, further, larger confirmatory studies are needed.
Collapse
Affiliation(s)
- Giles Major
- Nottingham Digestive Diseases Centre and NIHR Nottingham Biomedical Research Centre at Nottingham University Hospitals NHS Trust, the University of Nottingham, Nottingham, Notts, UK
| | - Lucy Bradshaw
- Nottingham Clinical Trials Unit (NCTU), University of Nottingham, Nottingham, UK
| | - Nafisa Boota
- Leicester Clinical Trials Unit, University of Leicester, Leicester, UK
| | - Kirsty Sprange
- Nottingham Clinical Trials Unit (NCTU), University of Nottingham, Nottingham, UK
| | - Mathew Diggle
- Clinical Microbiology Department, Nottingham University Hospitals NHS Trust, Nottingham, Nottinghamshire, UK
| | - Alan Montgomery
- Nottingham Clinical Trials Unit (NCTU), University of Nottingham, Nottingham, UK
| | - Aida Jawhari
- Clinical Microbiology Department, Nottingham University Hospitals NHS Trust, Nottingham, Nottinghamshire, UK
| | - Robin C Spiller
- Nottingham Digestive Diseases Centre and NIHR Nottingham Biomedical Research Centre at Nottingham University Hospitals NHS Trust, the University of Nottingham, Nottingham, Notts, UK
| | | |
Collapse
|
181
|
Formes H, Reinhardt C. The gut microbiota - a modulator of endothelial cell function and a contributing environmental factor to arterial thrombosis. Expert Rev Hematol 2019; 12:541-549. [PMID: 31159610 DOI: 10.1080/17474086.2019.1627191] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: There is emerging evidence linking the commensal gut microbiota with the development of cardiovascular disease and arterial thrombosis. In immunothrombosis, the host clotting system protects against the dissemination of invading microbes, not considering the huge number of microbes that interact with host physiology in a mutualistic fashion. Areas covered: Interestingly, recent research revealed that colonizing gut microbes profoundly influence host innate immune pathways that support arterial thrombus growth. The gut microbiota promotes arterial thrombus formation by enhancing the pro-adhesive capacity of the vascular endothelium, triggering hepatic von Willebrand factor synthesis and its release by Weibel-Palade body exocytosis, resulting in elevated von Willebrand factor levels and enhancing FVIII stability in plasma. Furthermore, the metabolic capacity of gut resident microbes promotes agonist-induced platelet activation and deposition. Here, we give an overview, with a focus on the vascular endothelium, on how this gut-resident microbial ecosystem contributes to arterial thrombus formation. Expert opinion: The gut microbiota and its metabolites not only act on agonist-induced platelet reactivity, but also influence the hepatic endothelial phenotype via remote signaling, facilitating arterial thrombus growth at the arterial injury site.
Collapse
Affiliation(s)
- Henning Formes
- a Center for Thrombosis and Hemostasis (CTH) , University Medical Center Mainz, Johannes Gutenberg University Mainz , Mainz , Germany
| | - Christoph Reinhardt
- a Center for Thrombosis and Hemostasis (CTH) , University Medical Center Mainz, Johannes Gutenberg University Mainz , Mainz , Germany.,b German Center for Cardiovascular Research (DZHK), University Medical Center Mainz, Partner Site RheinMain , Mainz , Germany
| |
Collapse
|
182
|
Lavelle A, Hill C. Gut Microbiome in Health and Disease: Emerging Diagnostic Opportunities. Gastroenterol Clin North Am 2019; 48:221-235. [PMID: 31046972 DOI: 10.1016/j.gtc.2019.02.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The gut microbiome is fundamental to human health and development. Altered microbiomes have been associated with many diseases. However, variation between individuals, environmental effects, and a lack of standardization across studies makes differentiation between health and disease challenging. Large-scale population cohorts in different countries will be required to match disease subjects with healthy controls, whereas standardized, reproducible pipelines for analysis are required to compare findings between studies. Despite this, several conditions have already demonstrated great promise for developing microbiome-based biomarkers as well as providing a gateway into integrated personalized medicine.
Collapse
Affiliation(s)
- Aonghus Lavelle
- Department of Medicine, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Colin Hill
- School of Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
183
|
Ziegler M, Han JH, Landsburg D, Pegues D, Reesey E, Gilmar C, Gorman T, Bink K, Moore A, Kelly BJ. Impact of Levofloxacin for the Prophylaxis of Bloodstream Infection on the Gut Microbiome in Patients With Hematologic Malignancy. Open Forum Infect Dis 2019; 6:ofz252. [PMID: 31281857 PMCID: PMC6602896 DOI: 10.1093/ofid/ofz252] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022] Open
Abstract
Background We evaluated the differential impact of levofloxacin administered for the prophylaxis of bloodstream infections compared with broad-spectrum beta-lactam (BSBL) antibiotics used for the treatment of neutropenic fever on the gut microbiome in patients with hematologic malignancy. Methods Stool specimens were collected from patients admitted for chemotherapy or stem cell transplant in the setting of the evaluation of diarrhea from February 2017 until November 2017. Microbiome characteristics were compared among those exposed to levofloxacin prophylaxis vs those who received BSBL antibiotics. Results Sixty patients were included, most with acute myeloid leukemia (42%) or multiple myeloma (37%). The gut microbiome of patients with BSBL exposure had significantly reduced Shannon's alpha diversity compared with those without (median [interquartile range {IQR}], 3.28 [1.73 to 3.71] vs 3.73 [3.14 to 4.31]; P = .01). However, those with levofloxacin exposure had increased alpha diversity compared with those without (median [IQR], 3.83 [3.32 to 4.36] vs 3.32 [2.35 to 4.02]; P = .03). Levofloxacin exposure was also associated with a trend toward lower risk of dominance of non-Bacteroidetes genera compared with those without levofloxacin exposure (3 [14%] vs 15 [38%]; P = .051). Conclusions The impact of antibiotics on the gut microbiome varies by class, and levofloxacin may disrupt the gut microbiome less than BSBLs in this patient population.
Collapse
Affiliation(s)
- Matthew Ziegler
- Division of Infectious Diseases, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jennifer H Han
- Division of Infectious Diseases, University of Pennsylvania, Philadelphia, Pennsylvania.,Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Healthcare Epidemiology, Infection Prevention and Control, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel Landsburg
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David Pegues
- Division of Infectious Diseases, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Healthcare Epidemiology, Infection Prevention and Control, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Emily Reesey
- Division of Infectious Diseases, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Cheryl Gilmar
- Department of Healthcare Epidemiology, Infection Prevention and Control, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Theresa Gorman
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kristen Bink
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amy Moore
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brendan J Kelly
- Division of Infectious Diseases, University of Pennsylvania, Philadelphia, Pennsylvania.,Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, Pennsylvania
| | | |
Collapse
|
184
|
Ganetsky A, Han JH, Hughes ME, Babushok DV, Frey NV, Gill SI, Hexner EO, Loren AW, Luger SM, Mangan JK, Martin ME, Smith J, Freyer CW, Gilmar C, Schuster M, Stadtmauer EA, Porter DL. Oral Vancomycin Prophylaxis Is Highly Effective in Preventing Clostridium difficile Infection in Allogeneic Hematopoietic Cell Transplant Recipients. Clin Infect Dis 2019; 68:2003-2009. [PMID: 30256954 PMCID: PMC6541731 DOI: 10.1093/cid/ciy822] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/20/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Clostridium difficile infection (CDI) is a leading cause of infectious complications in allogeneic hematopoietic cell transplant recipients (alloHCT). We sought to evaluate whether prophylactic oral vancomycin reduces the incidence of CDI in alloHCT recipients. METHODS We conducted a retrospective cohort study to examine the effectiveness of CDI prophylaxis with oral vancomycin, as compared to no prophylaxis, in 145 consecutive adult alloHCT recipients at the University of Pennsylvania between April 2015 and November 2016. Patients received oral vancomycin 125 mg twice daily, starting on admission and continuing until discharge. The primary outcome of interest was the association between oral vancomycin prophylaxis and CDI diagnosis. Secondary outcomes included graft-versus-host disease (GVHD) and relapse. RESULTS There were no cases of CDI in patients that received prophylaxis (0/90, 0%), whereas 11/55 (20%) patients who did not receive prophylaxis developed CDI (P < .001). Oral vancomycin prophylaxis was not associated with a higher risk of acute, grades 2-4 GVHD (subhazard ratio [sHR] 1.59; 95% confidence interval [CI] 0.88-2.89; P = .12), acute, grades 3-4 GVHD (sHR 0.65; 95% CI 0.25-1.66; P = .36), or acute, grades 2-4 gastrointestinal GVHD (sHR 1.95; 95% CI 0.93-4.07; P = .08) at day 180 post-transplant. No associations between oral vancomycin and relapse or survival were observed. CONCLUSIONS Prophylaxis with oral vancomycin is highly effective in preventing CDI in alloHCT recipients without increasing the risk of graft-versus-host disease or disease relapse. Further evaluation via a prospective study is warranted.
Collapse
Affiliation(s)
- Alex Ganetsky
- Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia
| | - Jennifer H Han
- Division of Infectious Diseases, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
- Department of Healthcare Epidemiology, Infection Prevention and Control, Hospital of the University of Pennsylvania, Philadelphia
| | - Mitchell E Hughes
- Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia
| | - Daria V Babushok
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Noelle V Frey
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Saar I Gill
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Elizabeth O Hexner
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Alison W Loren
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Selina M Luger
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - James K Mangan
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Mary Ellen Martin
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Jacqueline Smith
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Craig W Freyer
- Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia
| | - Cheryl Gilmar
- Department of Healthcare Epidemiology, Infection Prevention and Control, Hospital of the University of Pennsylvania, Philadelphia
| | - Mindy Schuster
- Division of Infectious Diseases, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Edward A Stadtmauer
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - David L Porter
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| |
Collapse
|
185
|
Flipse J, von Wintersdorff CJH, van Niekerk JM, Jamin C, van Tiel FH, Hasman H, van Alphen LB. Appearance of vanD-positive Enterococcus faecium in a tertiary hospital in the Netherlands: prevalence of vanC and vanD in hospitalized patients. Sci Rep 2019; 9:6949. [PMID: 31061446 PMCID: PMC6502811 DOI: 10.1038/s41598-019-42824-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 02/14/2019] [Indexed: 02/06/2023] Open
Abstract
Vancomycin-resistant enterococci (VRE) can rapidly spread through hospitals. Therefore, our hospital employs a screening program whereby rectal swabs are screened for the presence of vanA and vanB, and only PCR-positive broths are cultured on VRE selection agar. Early November 2016, a clinical vanA-/vanB-negative VRE isolate was detected in a vanA/vanB-screening-negative patient, giving the possibility that an undetected VRE might be spreading within our hospital. Whole-genome-sequencing of the isolate showed that resistance was vanD-mediated and core genome multilocus sequence typing showed it was a rare type: ST17/CT154. To determine the prevalence of vanA/B/C/D-carrying enterococci, we designed a real-time PCR for vanC1/2/3 and vanD and screened rectal swabs from 360 patients. vanD was found in 27.8% of the patients, yet culture demonstrated only E. faecium from vanA-positive broths and E. gallinarum from vanC1-positive broths. No vanD-positive VRE were found, limiting the possibility of nosocomial spread of this VRE. Moreover, the high prevalence of non-VRE vanD in rectal swabs makes it unfeasible to include the vanD PCR in our VRE screening. However, having validated the vanC1/2/3 and vanD PCRs allows us to rapidly check future vanA/B-negative VRE for the presence of vanC and vanD genes.
Collapse
Affiliation(s)
- Jacky Flipse
- Department of Medical Microbiology, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Center+, Maastricht, The Netherlands. .,Laboratory for Medical Microbiology and Infectious Diseases, Isala Clinics, Zwolle, The Netherlands.
| | - Christian J H von Wintersdorff
- Department of Medical Microbiology, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Julius M van Niekerk
- Department of Medical Microbiology, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Casper Jamin
- Department of Medical Microbiology, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Frank H van Tiel
- Department of Medical Microbiology, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Henrik Hasman
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Lieke B van Alphen
- Department of Medical Microbiology, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Center+, Maastricht, The Netherlands.
| |
Collapse
|
186
|
Han SH, Yi J, Kim JH, Lee S, Moon HW. Composition of gut microbiota in patients with toxigenic Clostridioides (Clostridium) difficile: Comparison between subgroups according to clinical criteria and toxin gene load. PLoS One 2019; 14:e0212626. [PMID: 30785932 PMCID: PMC6382146 DOI: 10.1371/journal.pone.0212626] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 02/06/2019] [Indexed: 12/19/2022] Open
Abstract
Data concerning the human microbiota composition during Clostridioides (Clostridium) difficile infection (CDI) using next-generation sequencing are still limited. We aimed to confirm key features indicating tcdB positive patients and compare the microbiota composition between subgroups based on toxin gene load (tcdB gene) and presence of significant diarrhea. Ninety-nine fecal samples from 79 tcdB positive patients and 20 controls were analyzed using 16S rRNA gene sequencing. Chao1 index for alpha diversity were calculated and principal coordinate analysis was performed for beta diversity using Quantitative Insights into Microbial Ecology (QIIME) pipeline. The mean relative abundance in each group was compared at phylum, family, and genus levels. There were significant alterations in alpha and beta diversity in tcdB positive patients (both colonizer and CDI) compared with those in the control. The mean Chao1 index of tcdB positive patients was significantly lower than the control group (P<0.001), whereas there was no significant difference between tcdB groups and between colonizer and CDI. There were significant differences in microbiota compositions between tcdB positive patients and the control at phylum, family, and genus levels. Several genera such as Phascolarctobacterium, Lachnospira, Butyricimonas, Catenibacterium, Paraprevotella, Odoribacter, and Anaerostipes were not detected in most CDI cases. We identified several changes in the microbiota of CDI that could be further evaluated as predictive markers. Microbiota differences between clinical subgroups of CDI need to be further studied in larger controlled studies.
Collapse
Affiliation(s)
- Sung-Hee Han
- BioCore Co. Ltd., Biotechnology, Yongin, Republic of Korea
| | - Joowon Yi
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Ji-Hoon Kim
- BioCore Co. Ltd., R&D Center, Seoul, Republic of Korea
| | - SangWon Lee
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hee-Won Moon
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
187
|
Bouza E, Cobo J, Almirante B, el Grupo de trabajo CLODIEXPAN. [Recommendations from a panel of experts on the usefulness of fidaxomicin for the treatment of infections caused by Clostridium difficile]. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2019; 32:50-59. [PMID: 30547500 PMCID: PMC6372963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Clostridium difficile infections have a high recurrence rate, which can complicate the prognosis of affected patients. It is therefore important to establish an early detection and an appropriate therapeutic strategy. The objective of this manuscript was to gather the opinion of an expert group about the predictive factors of poor progression, as well as when to use fidaxomicin in different groups of high-risk patients. METHODS A scientific committee of three experts in infectious diseases reviewed the most recent literature on the management of C. difficile infections, and the use of fidaxomicin. They developed a questionnaire of 23 items for consensus by 15 specialists in this type of infection using a modified Delphi method. RESULTS The consensus reached by the panelists was 91.3% in terms of agreement. The most important agreements were: recurrence is a risk criterion per se; fidaxomicin is effective and safe for the treatment of infections caused by C. difficile in critical patients, immunosuppressed patients, or patients with chronic renal failure; fidaxomicin is recommended from the first episode of infection to ensure maximum efficacy in patients with well-contrasted recurrence risk factors. CONCLUSIONS The experts consulted showed a high degree of agreement on topics related to the selection of patients with poorer prognosis, as well as on the use of fidaxomicin in groups of high-risk patients, either in the first line or in situations of recurrence.
Collapse
Affiliation(s)
- Emilio Bouza
- Hospital General Universitario Gregorio Marañón. Madrid
| | - Javier Cobo
- Servicio de Enfermedades infecciosas. Hospital Universitario Ramón y Cajal. Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS). Madrid
| | - Benito Almirante
- Servicio de Enfermedades Infecciosas. Hospital Universitario Vall d’Hebron. Barcelona
| | | |
Collapse
|
188
|
Vaughn BP, Kaiser T, Staley C, Hamilton MJ, Reich J, Graiziger C, Singroy S, Kabage AJ, Sadowsky MJ, Khoruts A. A pilot study of fecal bile acid and microbiota profiles in inflammatory bowel disease and primary sclerosing cholangitis. Clin Exp Gastroenterol 2019; 12:9-19. [PMID: 30666146 PMCID: PMC6330977 DOI: 10.2147/ceg.s186097] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Introduction Inflammatory bowel disease (IBD) is thought to arise from an abnormal immune response to the gut microbiota. IBD is associated with altered intestinal microbial community structure and functionality, which may contribute to inflammation and complications such as colon cancer and liver disease. Primary sclerosing cholangitis (PSC) is associated with IBD and markedly increases the risk of colon cancer. We hypothesized that secondary bile acids, which are products of microbial metabolism, are increased in PSC patients. Aim Here, we profiled the fecal bile acid composition and gut microbiota of participants with IBD and PSC, as well as healthy participants. Additionally, we tested the effects of vancomycin, a proposed treatment for PSC, on gut microbiota and fecal bile acid composition in participants with IBD and PSC. Methods Fecal samples were collected from patients with IBD, IBD/PSC and healthy controls and fecal bile acids and DNA for microbiota analysis were extracted. Fecal bile acids were averaged over a seven-day period. For subjects with IBD/PSC, oral vancomycin 500mg twice a day was administered and fecal samples were collected for up to eleven weeks. Results Participants with IBD and PSC had less fecal microbial diversity at baseline relative to controls. While there was some evidence of altered conversion of cholic acid to deoxycholic acid, no substantial differences were found in the fecal bile acid profiles of patients with IBD and PSC (n=7) compared to IBD alone (n=8) or healthy controls (n=8). Oral vancomycin was a potent inhibitor of secondary bile acid production in participants with IBD and PSC, particularly deoxycholic acid, although no changes in liver biochemistry patterns were noted over a two week period. Conclusion In this pilot study, bile acid profiles were overall similar among patients with IBD and PSC, IBD alone, and healthy controls. Microbiota diversity was reduced in those with PSC and IBD compared to IBD alone or healthy controls.
Collapse
Affiliation(s)
- Byron P Vaughn
- Inflammatory Bowel Program, Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota, Minneapolis, MN, USA,
| | - Thomas Kaiser
- BioTechnology Institute, University of Minnesota, St Paul, MN, USA.,Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Christopher Staley
- BioTechnology Institute, University of Minnesota, St Paul, MN, USA.,Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | | | - Jon Reich
- Inflammatory Bowel Program, Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota, Minneapolis, MN, USA,
| | - Carolyn Graiziger
- Inflammatory Bowel Program, Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota, Minneapolis, MN, USA,
| | | | - Amanda J Kabage
- Inflammatory Bowel Program, Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota, Minneapolis, MN, USA,
| | - Michael J Sadowsky
- BioTechnology Institute, University of Minnesota, St Paul, MN, USA.,Department of Soil, Water, and Climate, University of Minnesota, St Paul, MN, USA.,Department of Plant and Microbial Biology, University of Minnesota, St Paul, MN, USA
| | - Alexander Khoruts
- Inflammatory Bowel Program, Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota, Minneapolis, MN, USA, .,BioTechnology Institute, University of Minnesota, St Paul, MN, USA
| |
Collapse
|
189
|
Keith JW, Pamer EG. Enlisting commensal microbes to resist antibiotic-resistant pathogens. J Exp Med 2019; 216:10-19. [PMID: 30309968 PMCID: PMC6314519 DOI: 10.1084/jem.20180399] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/31/2018] [Accepted: 09/11/2018] [Indexed: 12/13/2022] Open
Abstract
The emergence of antibiotic-resistant bacterial pathogens is an all-too-common consequence of antibiotic use. Although antibiotic resistance among virulent bacterial pathogens is a growing concern, the highest levels of antibiotic resistance occur among less pathogenic but more common bacteria that are prevalent in healthcare settings. Patient-to-patient transmission of these antibiotic-resistant bacteria is a perpetual concern in hospitals. Many of these resistant microbes, such as vancomycin-resistant Enterococcus faecium and carbapenem-resistant Klebsiella pneumoniae, emerge from the intestinal lumen and invade the bloodstream of vulnerable patients, causing disseminated infection. These infections are associated with preceding antibiotic administration, which changes the intestinal microbiota and compromises resistance to colonization by antibiotic-resistant bacteria. Recent and ongoing studies are increasingly defining commensal bacterial species and the inhibitory mechanisms they use to prevent infection. The use of next-generation probiotics derived from the intestinal microbiota represents an alternative approach to prevention of infection by enriching colonization with protective commensal species, thereby reducing the density of antibiotic-resistant bacteria and also reducing patient-to-patient transmission of infection in healthcare settings.
Collapse
Affiliation(s)
- James W Keith
- Immunology Program, Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, New York, NY
| | - Eric G Pamer
- Immunology Program, Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, New York, NY
- Sloan Kettering Institute, New York, NY
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
190
|
Voigt AM, Faerber HA, Wilbring G, Skutlarek D, Felder C, Mahn R, Wolf D, Brossart P, Hornung T, Engelhart S, Exner M, Schmithausen RM. The occurrence of antimicrobial substances in toilet, sink and shower drainpipes of clinical units: A neglected source of antibiotic residues. Int J Hyg Environ Health 2019; 222:455-467. [PMID: 30622005 DOI: 10.1016/j.ijheh.2018.12.013] [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: 08/30/2018] [Revised: 12/22/2018] [Accepted: 12/31/2018] [Indexed: 12/18/2022]
Abstract
Antibiotics represent one of the most important drug groups used in the management of bacterial infections in humans and animals. Due to the increasing problem of antibiotic resistance, assurance of the antibacterial effectiveness of these substances has moved into the focus of public health. The reduction in antibiotic residues in wastewater and the environment may play a decisive role in the development of increasing rates of antibiotic resistance. The present study examines the wastewater of 31 patient rooms of various German clinics for possible residues of antibiotics, as well as the wastewater of five private households as a reference. To the best of our knowledge, this study shows for the first time that in hospitals with high antibiotic consumption rates, residues of these drugs can be regularly detected in toilets, sink siphons and shower drains at concentrations ranging from 0.02 μg·L-1 to a maximum of 79 mg·L-1. After complete flushing of the wastewater siphons, antibiotics are no longer detectable, but after temporal stagnation, the concentration of the active substances in the water phases of respective siphons increases again, suggesting that antibiotics persist through the washing process in biofilms. This study demonstrates that clinical wastewater systems offer further possibilities for the optimization of antibiotic resistance surveillance.
Collapse
Affiliation(s)
- A M Voigt
- Institute for Hygiene and Public Health, University Hospital Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - H A Faerber
- Institute for Hygiene and Public Health, University Hospital Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany.
| | - G Wilbring
- Institute for Hygiene and Public Health, University Hospital Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - D Skutlarek
- Institute for Hygiene and Public Health, University Hospital Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - C Felder
- Institute for Hygiene and Public Health, University Hospital Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - R Mahn
- Medical Clinic, Department of Haematology and Oncology, Centre for Integrated Oncology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - D Wolf
- Medical Clinic, Department of Haematology and Oncology, Centre for Integrated Oncology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany; University Clinic V, Dpt. Hematology and Oncology, Medical University Innsbruck, Anichstraße 35, A-6020 Innsbruck, Austria
| | - P Brossart
- Medical Clinic, Department of Haematology and Oncology, Centre for Integrated Oncology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - T Hornung
- Clinic and Polyclinic for Dermatology and Allergology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - S Engelhart
- Institute for Hygiene and Public Health, University Hospital Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - M Exner
- Institute for Hygiene and Public Health, University Hospital Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - R M Schmithausen
- Institute for Hygiene and Public Health, University Hospital Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| |
Collapse
|
191
|
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
|
192
|
Djukovic A, Garcia-Garcera M, Martínez-Paredes E, Isaac S, Artacho A, Martínez J, Ubeda C. Gut colonization by a novel Clostridium species is associated with the onset of epizootic rabbit enteropathy. Vet Res 2018; 49:123. [PMID: 30572930 PMCID: PMC6302431 DOI: 10.1186/s13567-018-0617-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/24/2018] [Indexed: 02/08/2023] Open
Abstract
Epizootic rabbit enteropathy (ERE) represents one of the most devastating diseases affecting rabbit farms. Previous studies showing transmissibility of disease symptoms through oral inoculation of intestinal contents from sick animals suggested a bacterial infectious origin for ERE. However, no etiological agent has been identified yet. On the other hand, ERE is associated with major changes in intestinal microbial communities, pinpointing dysbiosis as an alternative cause for the disease. To better understand the role of intestinal bacteria in ERE development, we have performed a prospective longitudinal study in which intestinal samples collected from the same animals before, during and after disease onset were analyzed using high-throughput sequencing. Changes in hundreds of bacterial groups were detected after the initiation of ERE. In contrast, before ERE onset, the microbiota from rabbits that developed ERE did not differ from those that remained healthy. Notably, an expansion of a single novel Clostridium species (Clostridium cuniculi) was detected the day of ERE onset. C. cuniculi encodes several putative toxins and it is phylogenetically related to the two well-characterized pathogens C. botulinum and C. perfringens. Our results are consistent with a bacterial infectious origin of ERE and discard dysbiosis as the initial trigger of the disease. Although experimental validation is required, results derived from sequencing analysis, propose a key role of C. cuniculi in ERE initiation.
Collapse
Affiliation(s)
- Ana Djukovic
- Departamento de Genómica y Salud, Centro Superior de Investigación en Salud Pública - FISABIO, Avenida de Cataluña, 21, 46020, Valencia, Valencia, Spain
| | - Marc Garcia-Garcera
- Department of Fundamental Microbiology, University of Lausanne, 1006, Lausanne, Switzerland
| | - Eugenio Martínez-Paredes
- Instituto de Ciencia y Tecnología Animal, Universitat Politècnica de Valencia, Camino de Vera, s/n., 46022, Valencia, Valencia, Spain
| | - Sandrine Isaac
- Departamento de Genómica y Salud, Centro Superior de Investigación en Salud Pública - FISABIO, Avenida de Cataluña, 21, 46020, Valencia, Valencia, Spain
| | - Alejandro Artacho
- Departamento de Genómica y Salud, Centro Superior de Investigación en Salud Pública - FISABIO, Avenida de Cataluña, 21, 46020, Valencia, Valencia, Spain
| | - Jorge Martínez
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Bellaterra, 08193, Cerdanyola del Vallés, Spain
| | - Carles Ubeda
- Departamento de Genómica y Salud, Centro Superior de Investigación en Salud Pública - FISABIO, Avenida de Cataluña, 21, 46020, Valencia, Valencia, Spain. .,CIBER en Epidemiología y Salud Pública, 28029, Madrid, Spain.
| |
Collapse
|
193
|
Neuman H, Forsythe P, Uzan A, Avni O, Koren O. Antibiotics in early life: dysbiosis and the damage done. FEMS Microbiol Rev 2018; 42:489-499. [PMID: 29945240 DOI: 10.1093/femsre/fuy018] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 06/23/2018] [Indexed: 12/21/2022] Open
Abstract
Antibiotics are the most common type of medication prescribed to children, including infants, in the Western world. While use of antibiotics has transformed previously lethal infections into relatively minor diseases, antibiotic treatments can have adverse effects as well. It has been shown in children, adults and animal models that antibiotics dramatically alter the gut microbial composition. Since the gut microbiota plays crucial roles in immunity, metabolism and endocrinology, the effects of antibiotics on the microbiota may lead to further health complications. In this review, we present an overview of the effects of antibiotics on the microbiome in children, and correlate them to long-lasting complications of obesity, behavior, allergies, autoimmunity and other diseases.
Collapse
Affiliation(s)
- Hadar Neuman
- Faculty of Medicine, Bar Ilan University, Henrietta Szold 8, Safed 13115, Israel.,Ziv Medical Center, Derech HaRambam St., Safed 13100 Israel.,Zefat Academic College, Jerusalem St. 11, Safed 13206, Israel
| | - Paul Forsythe
- McMaster Brain-Body Institute, St. Joseph's Healthcare Hamilton, L8N 4A6 Hamilton, Ontario, Canada.,Firestone Institute for Respiratory Health and Department of Medicine, 50 Charlton Avenue East, McMaster University, L8N 4A6 Hamilton, Ontario, Canada
| | - Atara Uzan
- Faculty of Medicine, Bar Ilan University, Henrietta Szold 8, Safed 13115, Israel
| | - Orly Avni
- Faculty of Medicine, Bar Ilan University, Henrietta Szold 8, Safed 13115, Israel
| | - Omry Koren
- Faculty of Medicine, Bar Ilan University, Henrietta Szold 8, Safed 13115, Israel
| |
Collapse
|
194
|
Abstract
Radical changes in the composition, diversity and metabolic activity of gut microbiome in critically ill patients most probably affect adversely the outcome of treatment. Microbiota dysfunction may be a predictor and presumably the main cause of infectious complications and sepsis. Clinicists use objective scales for evaluation of patient condition severity including specific parameters of disorders of organs and systems; however, microbiota function is not considered specific and, hence, not evaluated. Technical capabilities of the recent decade have allowed characterizing the intestinal microbiota and that helped understanding the ongoing processes. The authors have analyzed data about the role of intestinal microbiota as a metabolic 'reactor' during critical states, possible complications related to misbalance of 'harmful' and 'beneficial' bacteria, and examined potential of a targeted therapy aimed directly at correction of intestinal microbiota. Search for papers was carried out using Scopus and Web of Science databases 2001 to 2018 years: (Gut Microbiota) AND (Critically ill OR Intensive care unit), key words taken for the search were: intestinal microbiota, metabolism, sepsis, antibiotics, critically ill patients, multiple organ failure. A number of questions in understanding of the interaction between gut microbiome and host remain open. It is necessary to take into account interference of microbial metabolism while assessing metabolome of patients with sepsis. Among low-molecular compounds found in blood of sepsis patients, special attention should be paid to molecules that can be classified as ‘common metabolites’ of humans and bacteria, for example, degradation products of aromatic compounds, which many-fold rise in blood of septic patients. It is necessary to take into consideration and experimentally model changes in the human internal environment, which occur during radical transformation of microbiome in critically ill patients. Such approach brings in new prospects for objective monitoring of diseases by evaluating metabolic profile at a particular moment of time based on integral indices reflecting the status of microbiome/metabolome system, which will supply new targets for therapeutic intervention in future.
Collapse
Affiliation(s)
- E. A. Chernevskaya
- V. A. Negovsky Research Institute of General Reanimatology, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology
| | - N. V. Beloborodova
- V. A. Negovsky Research Institute of General Reanimatology, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology
| |
Collapse
|
195
|
Coleman M, Elkins C, Gutting B, Mongodin E, Solano-Aguilar G, Walls I. Microbiota and Dose Response: Evolving Paradigm of Health Triangle. RISK ANALYSIS : AN OFFICIAL PUBLICATION OF THE SOCIETY FOR RISK ANALYSIS 2018; 38:2013-2028. [PMID: 29900563 DOI: 10.1111/risa.13121] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 01/31/2018] [Accepted: 03/17/2018] [Indexed: 06/08/2023]
Abstract
SRA Dose-Response and Microbial Risk Analysis Specialty Groups jointly sponsored symposia that addressed the intersections between the "microbiome revolution" and dose response. Invited speakers presented on innovations and advances in gut and nasal microbiota (normal microbial communities) in the first decade after the Human Microbiome Project began. The microbiota and their metabolites are now known to influence health and disease directly and indirectly, through modulation of innate and adaptive immune systems and barrier function. Disruption of healthy microbiota is often associated with changes in abundance and diversity of core microbial species (dysbiosis), caused by stressors including antibiotics, chemotherapy, and disease. Nucleic-acid-based metagenomic methods demonstrated that the dysbiotic host microbiota no longer provide normal colonization resistance to pathogens, a critical component of innate immunity of the superorganism. Diverse pathogens, probiotics, and prebiotics were considered in human and animal models (in vivo and in vitro). Discussion included approaches for design of future microbial dose-response studies to account for the presence of the indigenous microbiota that provide normal colonization resistance, and the absence of the protective microbiota in dysbiosis. As NextGen risk analysis methodology advances with the "microbiome revolution," a proposed new framework, the Health Triangle, may replace the old paradigm based on the Disease Triangle (focused on host, pathogen, and environment) and germophobia. Collaborative experimental designs are needed for testing hypotheses about causality in dose-response relationships for pathogens present in our environments that clearly compete in complex ecosystems with thousands of bacterial species dominating the healthy superorganism.
Collapse
|
196
|
Goldstein J, Levy C. Novel and emerging therapies for cholestatic liver diseases. Liver Int 2018; 38:1520-1535. [PMID: 29758112 DOI: 10.1111/liv.13880] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 05/01/2018] [Indexed: 02/06/2023]
Abstract
While bile acids are important for both digestion and signalling, hydrophobic bile acids can be harmful, especially when in high concentrations. Mechanisms for the protection of cholangiocytes against bile acid cytotoxicity include negative feedback loops via farnesoid X nuclear receptor (FXR) activation, the bicarbonate umbrella, cholehepatic shunting and anti-inflammatory signalling, among others. By altering or overwhelming these defence mechanisms, cholestatic diseases such as primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC) can further progress to biliary cirrhosis, end-stage liver disease and death or liver transplantation. While PBC is currently treated with ursodeoxycholic acid (UDCA) and obeticholic acid (OCA), many fail treatment, and we have yet to find an effective therapy for PSC. Novel therapies under evaluation target nuclear and surface receptors including FXR, transmembrane G-protein-coupled receptor 5 (TGR5), peroxisome proliferator-activated receptor (PPAR) and pregnane X receptor (PXR). Modulation of these receptors leads to altered bile composition, decreased cytotoxicity, decreased inflammation and improved metabolism. This review summarizes our current understanding of the role of bile acids in the pathophysiology of cholestatic liver diseases, presents the rationale for already approved medical therapies and discusses novel pharmacologic therapies under investigation.
Collapse
Affiliation(s)
- Jordan Goldstein
- Division of Internal Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Cynthia Levy
- Division of Hepatology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
197
|
Harris VC, Haak BW, Handley SA, Jiang B, Velasquez DE, Hykes BL, Droit L, Berbers GAM, Kemper EM, van Leeuwen EMM, Boele van Hensbroek M, Wiersinga WJ. Effect of Antibiotic-Mediated Microbiome Modulation on Rotavirus Vaccine Immunogenicity: A Human, Randomized-Control Proof-of-Concept Trial. Cell Host Microbe 2018; 24:197-207.e4. [PMID: 30092197 PMCID: PMC11514417 DOI: 10.1016/j.chom.2018.07.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/21/2018] [Accepted: 07/04/2018] [Indexed: 11/26/2022]
Abstract
Rotavirus vaccines (RVV) protect against childhood gastroenteritis caused by rotavirus (RV) but have decreased effectiveness in low- and middle-income settings. This proof-of-concept, randomized-controlled, open-label trial tested if microbiome modulation can improve RVV immunogenicity. Healthy adults were randomized and administered broad-spectrum (oral vancomycin, ciprofloxacin, metronidazole), narrow-spectrum (vancomycin), or no antibiotics and then vaccinated with RVV, 21 per group per protocol. Baseline anti-RV IgA was high in all subjects. Although antibiotics did not alter absolute anti-RV IgA titers, RVV immunogenicity was boosted at 7 days in the narrow-spectrum group. Further, antibiotics increased fecal shedding of RV while also rapidly altering gut bacterial beta diversity. Beta diversity associated with RVV immunogenicity boosting at day 7 and specific bacterial taxa that distinguish RVV boosters and RV shedders were identified. Despite the negative primary endpoint, this study demonstrates that microbiota modification alters the immune response to RVV and supports further exploration of microbiome manipulation to improve RVV immunogenicity.
Collapse
Affiliation(s)
- Vanessa C Harris
- Amsterdam UMC, University of Amsterdam, Department of Medicine, Division of Infectious Diseases and Center for Experimental and Molecular Medicine (CEMM), 1105 AZ, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Department of Global Health - Amsterdam Institute for Global Health and Development (AIGHD), 1105 AZ, Amsterdam, the Netherlands.
| | - Bastiaan W Haak
- Amsterdam UMC, University of Amsterdam, Department of Medicine, Division of Infectious Diseases and Center for Experimental and Molecular Medicine (CEMM), 1105 AZ, Amsterdam, the Netherlands
| | - Scott A Handley
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Baoming Jiang
- Division of Viral Diseases, Center for Disease Control and Prevention (CDC), Atlanta, GA 30329, USA
| | - Daniel E Velasquez
- Division of Viral Diseases, Center for Disease Control and Prevention (CDC), Atlanta, GA 30329, USA
| | - Barry L Hykes
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Lindsay Droit
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Guy A M Berbers
- Center for Infectious Disease Control, Netherlands National Institute for Public Health and the Environment (RIVM), 3721 MA, Bilthoven, the Netherlands
| | - Elles Marleen Kemper
- Amsterdam UMC, University of Amsterdam, Department of Pharmacy, 1105 AZ, Amsterdam, the Netherlands
| | - Ester M M van Leeuwen
- Amsterdam UMC, University of Amsterdam, Department of Experimental Immunology, 1105 AZ, Amsterdam, the Netherlands
| | - Michael Boele van Hensbroek
- Amsterdam UMC, University of Amsterdam, Department of Global Health - Amsterdam Institute for Global Health and Development (AIGHD), 1105 AZ, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Emma Children's Hospital, 1105 AZ, Amsterdam, the Netherlands
| | - Willem Joost Wiersinga
- Amsterdam UMC, University of Amsterdam, Department of Medicine, Division of Infectious Diseases and Center for Experimental and Molecular Medicine (CEMM), 1105 AZ, Amsterdam, the Netherlands
| |
Collapse
|
198
|
Thorpe CM, Kane AV, Chang J, Tai A, Vickers RJ, Snydman DR. Enhanced preservation of the human intestinal microbiota by ridinilazole, a novel Clostridium difficile-targeting antibacterial, compared to vancomycin. PLoS One 2018; 13:e0199810. [PMID: 30071046 PMCID: PMC6071993 DOI: 10.1371/journal.pone.0199810] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/09/2018] [Indexed: 12/21/2022] Open
Abstract
Ridinilazole, a novel targeted antibacterial being developed for the treatment of C. difficile infection (CDI) and prevention of recurrence, was shown in a recent Phase 2 study to be superior to vancomycin with regard to the primary efficacy measure, sustained clinical response (SCR), with the superiority being driven primarily by marked reductions in the rates of CDI recurrence within 30 days. Tolerability of ridinilazole was comparable to that of vancomycin. The current nested cohort study compared the effects of ridinilazole and vancomycin on fecal microbiota during and after treatment among participants in the Phase 2 study. Changes in the microbiota were assessed using qPCR and high-throughput sequencing on participants' stools collected at multiple time-points (baseline [Day 1], Day 5, end-of-treatment [EOT; Day 10], Day 25, end-of-study [EOS; Day 40], and at CDI recurrence). qPCR analyses showed profound losses of Bacteroides, C. coccoides, C. leptum, and Prevotella groups at EOT with vancomycin treatment, while ridinilazole-treated participants had a modest decrease in C. leptum group levels at EOT, with levels recovering by Day 25. Vancomycin-treated participants had a significant increase in the Enterobacteriaceae group, with this increase persisting beyond EOT. At EOT, alpha diversity decreased with both antibiotics, though to a significantly lesser extent with ridinilazole (p <0.0001). Beta diversity analysis showed a significantly larger weighted Unifrac distance from baseline-to-EOT with vancomycin. Taxonomically, ridinilazole had a markedly narrower impact, with modest reductions in relative abundance in Firmicutes taxa. Microbiota composition returned to baseline sooner with ridinilazole than with vancomycin. Vancomycin treatment resulted in microbiome-wide changes, with significant reductions in relative abundances of Firmicutes, Bacteroidetes, Actinobacteria, and a profound increase in abundance of Proteobacteria. These findings demonstrate that ridinilazole is significantly less disruptive to microbiota than vancomycin, which may contribute to the reduced CDI recurrence observed in the Phase 2 study.
Collapse
Affiliation(s)
- Cheleste M. Thorpe
- Tufts Medical Center, Boston, MA, United States of America
- Tufts University School of Medicine, Boston, MA, United States of America
| | - Anne V. Kane
- Tufts Medical Center, Boston, MA, United States of America
| | - Justin Chang
- Tufts University School of Medicine, Boston, MA, United States of America
| | - Albert Tai
- Tufts University School of Medicine, Boston, MA, United States of America
| | | | - David R. Snydman
- Tufts Medical Center, Boston, MA, United States of America
- Tufts University School of Medicine, Boston, MA, United States of America
| |
Collapse
|
199
|
Seekatz AM, Bassis CM, Fogg L, Moore NM, Rhee Y, Lolans K, Weinstein RA, Lin MY, Young VB, Hayden MK. Gut Microbiota and Clinical Features Distinguish Colonization With Klebsiella pneumoniae Carbapenemase-Producing Klebsiella pneumoniae at the Time of Admission to a Long-term Acute Care Hospital. Open Forum Infect Dis 2018; 5:ofy190. [PMID: 30151415 PMCID: PMC6101546 DOI: 10.1093/ofid/ofy190] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 07/27/2018] [Indexed: 12/29/2022] Open
Abstract
Background Identification of gut microbiota features associated with antibiotic-resistant bacterial colonization may reveal new infection prevention targets. Methods We conducted a matched, case–control study of long-term acute care hospital (LTACH) patients to identify gut microbiota and clinical features associated with colonization by Klebsiella pneumoniae carbapenemase-producing Klebsiella pneumoniae (KPC-Kp), an urgent antibiotic resistance threat. Fecal or rectal swab specimens were collected and tested for KPC-Kp; 16S rRNA gene-based sequencing was performed. Comparisons were made between cases and controls in calibration and validation subsamples using microbiota similarity indices, logistic regression, and unit-weighted predictive models. Results Case (n = 32) and control (n = 99) patients had distinct fecal microbiota communities, but neither microbiota diversity nor inherent clustering into community types distinguished case and control specimens. Comparison of differentially abundant operational taxonomic units (OTUs) revealed 1 OTU associated with case status in both calibration (n = 51) and validation (n = 80) subsamples that matched the canonical KPC-Kp strain ST258. Permutation analysis using the presence or absence of OTUs and hierarchical logistic regression identified 2 OTUs (belonging to genus Desulfovibrio and family Ruminococcaceae) associated with KPC-Kp colonization. Among clinical variables, the presence of a decubitus ulcer alone was independently and consistently associated with case status. Combining the presence of the OTUs Desulfovibrio and Ruminococcaceae with decubitus ulcer increased the likelihood of KPC-Kp colonization to >38% in a unit-weighted predictive model. Conclusions We identified microbiota and clinical features that distinguished KPC-Kp gut colonization in LTACH patients, a population particularly susceptible to KPC-Kp infection. These features may warrant further investigation as markers of risk for KPC-Kp colonization.
Collapse
Affiliation(s)
- Anna M Seekatz
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Christine M Bassis
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Louis Fogg
- Department of Nursing, Rush College of Nursing, Chicago, Illinois
| | | | - Yoona Rhee
- Division of Infectious Diseases, Department of Internal Medicine, Rush Medical College, Chicago, Illinois
| | | | - Robert A Weinstein
- Division of Infectious Diseases, Department of Internal Medicine, Rush Medical College, Chicago, Illinois.,Cook County Health and Hospitals System, Chicago, Illinois
| | - Michael Y Lin
- Division of Infectious Diseases, Department of Internal Medicine, Rush Medical College, Chicago, Illinois
| | - Vincent B Young
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Mary K Hayden
- Department of Pathology.,Division of Infectious Diseases, Department of Internal Medicine, Rush Medical College, Chicago, Illinois
| | | |
Collapse
|
200
|
Protection from chemotherapy- and antibiotic-mediated dysbiosis of the gut microbiota by a probiotic with digestive enzymes supplement. Oncotarget 2018; 9:30919-30935. [PMID: 30112118 PMCID: PMC6089397 DOI: 10.18632/oncotarget.25778] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/05/2018] [Indexed: 01/20/2023] Open
Abstract
There are numerous downstream consequences of marketed drugs like antineoplastic agents on the gut microbiome, an effect that is suggested to contribute to adverse event profiles and may also influence drug responses. In cancer, progress is needed toward modulation of the host microbiome to prevent off-target side effects of drugs such as gastrointestinal mucositis that result from gut dysbiosis. The objective of this study was evaluation of the bioactivity of a supplement consisting of capsules with a blend of 9 probiotic organisms of the genera Lactobacillus and Bifidobacterium plus 10 digestive enzymes, in protecting the human gastrointestinal tract from chemotherapy and an antibiotic. We used the Simulator of Human Intestinal Microbial Ecosystem (SHIME) model, an in vitro model of a stable colon microbiota, and introduced 5-fluorouracil (5-FU) and vancomycin as microbiome-disrupting drugs. The probiotic with digestive enzymes supplement, added in capsules at in vivo doses, improved fermentation activity in the colon reactors and accelerated the recovery of microbial populations following 5-FU/vancomycin treatment. The supplement restored the Bacteroidetes to Firmicutes ratios in the colon reactors, increased the diversity of microbiota, and induced the production of microbial metabolites that elicited anti-inflammatory cytokines in an in vitro model of intestinal inflammation. In the proximal colon, preventative administration of the supplement resulted in full recovery of the gut microbial community after cessation of 5-FU and vancomycin treatment. These results identify a probiotic with digestive enzymes formulation that protects against drug-induced gut dysbiosis, highlighting its potential utility as a component of routine cancer care.
Collapse
|