Characterizations of Clinical Isolates of Clostridium difficile by Toxin. Genotypes and by Susceptibility to 12 Antimicrobial Agents, Including

Similar documents
Should we test Clostridium difficile for antimicrobial resistance? by author

AAC Revised. Activity of a Novel Cyclic Lipopeptide, CB-183,315 Against Resistant Clostridium difficile

In vitro activity of surotomycin against contemporary clinical isolates of toxigenic Clostridium difficile strains obtained in Spain

Overview of C. difficile infections. Kurt B. Stevenson, MD MPH Professor Division of Infectious Diseases

Reply to Fabre et. al

Trends in Susceptibility of Vancomycin-resistant Enterococcus. faecium to Tigecycline, Daptomycin, and Linezolid and

Comparison of Supplemented Brucella Agar and Modified Clostridium difficile Agar for Antimicrobial Susceptibility Testing of Clostridium difficile

In vitro susceptibility to 17 antimicrobials of clinical Clostridium difficile isolates collected in in Sweden

In Vitro Activities of Linezolid against Clinical Isolates of ACCEPTED

on February 12, 2018 by guest

Comparison of antimicrobial susceptibility among Clostridium difficile isolated from an integrated human and swine population in Texas

Clostridium difficile

Clostridium difficile Infection Prevention. Basics of Infection Prevention 2-Day Mini-Course 2012

Annual Report: Table 1. Antimicrobial Susceptibility Results for 2,488 Isolates of S. pneumoniae Collected Nationally, 2005 MIC (µg/ml)

Inappropriate Use of Antibiotics and Clostridium difficile Infection. Jocelyn Srigley, MD, FRCPC November 1, 2012

Clinical and microbiologic characteristics of tcdanegative variant clostridium difficile infections

Learning Objectives 6/1/18

Clostridium Difficile Primer: Disease, Risk, & Mitigation

Please distribute a copy of this information to each provider in your organization.

Principles of Antimicrobial Therapy

Epidemiology of Clostridium difficile infections in a tertiary-care hospital in Korea

Healthcare-associated Infections Annual Report December 2018

against Clinical Isolates of Gram-Positive Bacteria

(c) 2016, Freeman et al. This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license.

Ed J. Kuijper National Reference Laboratory for Clostridium difficile Leiden University Medical Center

Antimicrobial susceptibility testing of Clostridium difficile using EUCAST epidemiological cut-off values and disk diffusion correlates

Incidence of hospital-acquired Clostridium difficile infection in patients at risk

Molecular characterization and antimicrobial susceptibility patterns of Clostridium difficile strains isolated from hospitals in south-east Scotland

Antimicrobial Susceptibility of Clinical Isolates of Bacteroides fragilis Group Organisms Recovered from 2009 to 2012 in a Korean Hospital

Received: February 29, 2008 Revised: July 22, 2008 Accepted: August 4, 2008

Clinical Spectrum of Disease. Clinical Features. Risk Factors. Risk of CDAD According to Antibiotic Class. Fluoroquinolones as Risk Factor for CDAD

Does Screening for MRSA Colonization Have A Role In Healthcare-Associated Infection Prevention Programs?

Impact of an intervention to control Clostridium difficile infection on hospital- and community-onset disease; an interrupted time series analysis

Antibiotic Reference Laboratory, Institute of Environmental Science and Research Limited (ESR); August 2017

Association of tcda+/tcdb+ Clostridium difficile Genotype with Emergence of Multidrug-Resistant Strains Conferring Metronidazole Resistant Phenotype

Marc Decramer 3. Respiratory Division, University Hospitals Leuven, Leuven, Belgium

Antimicrobial Resistance Strains

Review Article Clostridium difficile Infections: A Global Overview of Drug Sensitivity and Resistance Mechanisms

Clostridium difficile Colitis

Healthcare-associated Infections Annual Report

SESSION XVI NEW ANTIBIOTICS

Dynamic Drug Combination Response on Pathogenic Mutations of Staphylococcus aureus

EDUCATIONAL COMMENTARY - Methicillin-Resistant Staphylococcus aureus: An Update

Clostridium difficile infection: The Present and the Future

Healthcare-associated Infections Annual Report March 2015

Methicillin-Resistant Staphylococcus aureus

Multidrug-Resistant Salmonella enterica in the Democratic Republic of the Congo (DRC)

What is new in 2011: Methods and breakpoints in relation to subcommittees and expert groups. by author. Gunnar Kahlmeter, Derek Brown

Tel: Fax:

Clostridium Difficile Infection (CDI) Alistair McGregor Hobart Pathology Royal Hobart Hospital TIPCU

Principles and Practice of Antimicrobial Susceptibility Testing. Microbiology Technical Workshop 25 th September 2013

11/2/2015. Update on the Treatment of Clostridium difficile Infections. Disclosure. Objectives

Int.J.Curr.Microbiol.App.Sci (2018) 7(8):

Clostridium difficile may be found in 1% to 3% of all

Antibiotic Updates: Part II

PK/PD to fight resistance

3/9/15. Disclosures. Salmonella and Fluoroquinolones: Where are we now? Salmonella Current Taxonomy. Salmonella spp.

Antimicrobial Cycling. Donald E Low University of Toronto

Moxifloxacin resistance is prevalent among Bacteroides and Prevotella species in Greece

Evaluating the Role of MRSA Nasal Swabs

C.difficile Re-emergence of an Old Pathogen

Section 10: Antimicrobial Stewardship and Clostridium difficile Infection: A Primer for the Infection Preventionist

Vancomycin for the Treatment of Clostridium difficile Infection: For Whom Is This Expensive Bullet Really Magic?

Is biocide resistance already a clinical problem?

Appropriate antimicrobial therapy in HAP: What does this mean?

Newsflash: Hospital Medicine JOHN C. CHRISTENSEN, MD FACP AMERICAN COLLEGE OF PHYSICIANS, UTAH CHAPTER SCIENTIFIC MEETING FEBRUARY 10, 2017

ESBL Producers An Increasing Problem: An Overview Of An Underrated Threat

Preventing Clostridium difficile Infection (CDI)

Proceedings of the 19th American Academy of Veterinary Pharmacology and Therapeutics Biennial Symposium

In vitro Activity Evaluation of Telavancin against a Contemporary Worldwide Collection of Staphylococcus. aureus. Rodrigo E. Mendes, Ph.D.

Antimicrobial susceptibility of Clostridium difficile isolated in Thailand

Antibiotics in vitro : Which properties do we need to consider for optimizing our therapeutic choice?

Preventing Clostridium difficile. July 13,

FM - Male, 38YO. MRSA nasal swab (+) Due to positive MRSA nasal swab test, patient will be continued on Vancomycin 1500mg IV q12 for MRSA treatment...

ORIGINAL ARTICLE. Focus Technologies, Inc., 1 Hilversum, The Netherlands, 2 Herndon, Virginia and 3 Franklin, Tennessee, USA

Title: Effect of fidaxomicin versus vancomycin on susceptibility to intestinal

Christiane Gaudreau* and Huguette Gilbert

Available online at journal homepage:

In vitro activity of tigecycline against methicillin-resistant Staphylococcus aureus, including livestock-associated strains

Towards Rational International Antibiotic Breakpoints: Actions from the European Committee on Antimicrobial Susceptibility Testing (EUCAST)

Antibiotic Stewardship in LTC What does this mean?

USA Product Label CLINTABS TABLETS. Virbac. brand of clindamycin hydrochloride tablets. ANADA # , Approved by FDA DESCRIPTION

Consequences of Antimicrobial Resistant Bacteria. Antimicrobial Resistance. Molecular Genetics of Antimicrobial Resistance. Topics to be Covered

MID 23. Antimicrobial Resistance. Consequences of Antimicrobial Resistant Bacteria. Molecular Genetics of Antimicrobial Resistance

The Journal of Veterinary Medical Science

Microbiology : antimicrobial drugs. Sheet 11. Ali abualhija

Safe Patient Care Keeping our Residents Safe Use Standard Precautions for ALL Residents at ALL times

Antimicrobial Resistance

Antimicrobial Resistance Acquisition of Foreign DNA

Antimicrobial stewardship: Quick, don t just do something! Stand there!

Antibiotic Stewardship in the Hospital Setting

Anaerobe bakterier og resistens. Ulrik Stenz Justesen Klinisk Mikrobiologisk Afdeling Odense Universitetshospital Odense, Denmark

Randall Singer, DVM, MPVM, PhD

Community-Associated C. difficile Infection: Think Outside the Hospital. Maria Bye, MPH Epidemiologist May 1, 2018

Background and Plan of Analysis

Guideline Updates Change is Inevitable Especially in Infectious Diseases!

Veterinary Microbiology

Cipro and the aorta Fluoroquinolone attack? Bulat A. Ziganshin, MD, PhD and John A. Elefteriades, MD, PhD (hon)

ESCMID Online Lecture Library. by author

Antimicrobial Susceptibility of Clinically Relevant Gram-Positive Anaerobic Cocci Collected over a Three-Year Period in the Netherlands

Transcription:

AAC Accepts, published online ahead of print on 16 April 2012 Antimicrob. Agents Chemother. doi:10.1128/aac.00191-12 Copyright 2012, American Society for Microbiology. All Rights Reserved. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 Characterizations of Clinical Isolates of Clostridium difficile by Toxin Genotypes and by Susceptibility to 12 Antimicrobial Agents, Including Fidaxomicin (OPT-80) and Rifaximin: A Multicenter Study in Taiwan Chun-Hsing Liao, 1 Wen-Chien Ko, 2 Jang-Jih Lu, 3 and Po-Ren Hsueh 4 * Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan 1 ; Department of Internal Medicine and Center for Infection Control, National Cheng Kung University Hospital and Medical College, Tainan, Taiwan 2 ; Department of Laboratory Medicine, Chang Gung Memorial Hospital, Chang Gung University Medical College, Lin-Ko, Taiwan 3 ; Departments of Laboratory Medicine and Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan 4 * Corresponding author. Mailing address for Po-Ren Hsueh: Departments of Laboratory Medicine and Internal Medicine, National Taiwan University Hospital, No. 7 Chung-Shan South Rd., Taipei 100, Taiwan. Phone: 886-2-23123456, ext. 65355. Fax: 886-2-23224263. E-mail: hsporen@ntu.edu.tw. 1

21 ABSTRACT 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 A total of 403 non-duplicate isolates of Clostridium difficile were collected at three major teaching hospitals representing northern, central, and southern Taiwan from January 2005 to December 2010. Of these 403 isolates, 170 (42.2%) were presumed to be nontoxigenic due to the absence of genes for toxins A or B or binary toxin. The remaining 233 (57.8%) isolates carried toxin A and B genes, and 39 (16.7%) of these also had binary toxin genes. The MIC 90 of all isolates for fidaxomicin and rifaximin was 0. 5 μg/ml (range, 0.015-0.5 μg/ml) and >128 μg/ml (range, 0.015->128 μg/ml), respectively. All isolates were susceptible to metronidazole (MIC 90 0. 5 μg/ml; range, 0.03-4 μg/ml). Two isolates had reduced susceptibility to vancomycin (MICs, 4 μg/ml). Only 13.6% of isolates were susceptible to clindamycin (MIC 2 μg/ml). Nonsusceptibility to moxifloxacin (n=81, 20.1%) was accompanied by single or multiple mutations in gyra and gyrb genes in all but eight of moxifloxacin-nonsusceptible isolates. Two previously unreported gyrb mutations might independently confer resistance (MIC, 16 μg/ml), Ser416 to Ala and Glu466 to Lys. Moxifloxacin-resistant isolates were cross-resistant to ciprofloxacin and levofloxacin, but some moxifloxacin-nonsusceptible isolates remained susceptible to gemifloxacin or nemonoxacin at 0.5 μg/ml. This study found the diversity of toxigenic and nontoxigenic strains of C. difficile found in the healthcare setting in Taiwan. All isolates tested were susceptible to metronidazole and vancomycin. Fidaxomicin exhibited potent in vitro activity against all isolates tested, while more than 10% of Taiwanese isolates with rifaximin MICs of 128 μg/ml needs more concerns. 2

43 INTRODUCTION 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 Clostridium difficile infection (CDI) is a major nosocomial threat and may surpass methicillin-resistant Staphylococcus aureus in some settings (28). Although the two most common therapies for CDI, metronidazole and vancomycin, are effective in resolving most cases (4, 7), there is concern that efficacy of metronidazole is declining in recent outbreaks and that overuse of vancomycin can lead to selection of vancomycin-resistant enterococci (2, 3, 7, 30, 40). Approximately 20-30% of patients have recurrence of CDI after successful treatment with metronidazole or vancomycin. In patients with multiple recurrences, tapered doses of vancomycin or use of a rifaximin chaser are sometimes effective (4, 7, 14, 15). Not all C. difficile strains are pathogenic. Toxigenic strains harbor genes encoded by the pathogenicity locus (PaLoc), including cdta encoding enterotoxin A and cdtb encoding enterotoxin B as well as a negative regulator of their expression, cdtc (9). Emergence of a particularly virulent strain since 2000 has accounted for increased mortality in outbreaks in Europe, Canada, and the United States (24, 27, 29, 32, 39). This strain, restriction endonuclease analysis group type BI/pulsed field gel electrophoresis type 1/PCR ribotype 027 (BI/NAP1/027), is characterized by its resistance to fluoroquinolones, mutations in the cdtc gene, and expression of an ADP-ribosylating binary toxin, encoded outside the PaLoc locus and not expressed in most toxigenic strains (31). Furthermore, the link between toxin profiles, antibiotypes (including clindamycin and quinolones), and epidemicity is an important given the emergence and epidemic spread of pathogenic strains of C. difficile (33). 63 3

64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 To date, BI/NAP1/027 has not been documented in Taiwan (5, 20, 25, 26). However, C. difficile clinical isolates resistant to fluoroquinolones have been found (26). Greater awareness in Taiwan in the last decade has prompted retrospective and prospective surveillance studies in some hospitals. Hsu et al reported an incidence of 8 cases per 1000 patient-days in Northern Taiwan during a 3-month period in 2003 (20). The same hospital conducted a 5-month prospective surveillance in high-risk units of the same hospital during 2010 and found a much lower incidence of 0.45 cases per 1000 patient-days after initiating an aggressive hand-washing program (5, 25). In a teaching hospital in Southern Taiwan over a 15-month period during 2007-2008, a very similar rate of 0.43 cases per 1000 patient-days was recorded, with a higher rate of 1.1 cases per 1000 patient-days in the intensive care unit (5). We recently reported the antibiotic susceptibility profiles and molecular epidemiology of 113 C. difficile isolates from two major teaching hospitals in Northern and Southern Taiwan (26). In the current study, we extend these results to the molecular and microbiological characterization of 403 isolates from three hospitals representing northern, central, and southern Taiwan. Susceptibility to clindamycin and major fluoroquinolones, a non-fluorinated quinolone (nemonoxacin), and to antibiotics used clinically against CDI are reported and compared to genotypes for PaLoc toxins A and B and binary toxin and mutations in the DNA gyrase A and B genes. We also included fidaxomicin, a macrocyclic antibiotic with high specificity for C. difficile and inhibitory activity toward C. difficile RNA polymerase, and another RNA polymerase inhibitor, rifaximin, in this study. 4

84 MATERIALS AND METHODS 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 Bacterial isolates. A total of 403 non-duplicate isolates of C. difficile, including 332 isolates from National Taiwan University Hospital (NTUH), 40 from National Cheng Kung University Hospital (NCKUH), and 31 from China Medical University from January 2005 to December 2010, were obtained for analysis. These isolates were recovered from stool specimens of patients with unexplained fever or concurrent gastrointestinal symptoms, such as diarrhea, abdominal discomfort or ileus. Not all patients were confirmed as having CDI by toxin assays. Antimicrobial susceptibility testing. Minimum inhibitory concentrations (MICs) of the 403 isolates to 12 antimicrobial agents were determined using the agar dilution method recommended by the Clinical and Laboratory Standards Institute (CLSI) (6), with the exception of daptomycin. An inoculum of 105 CFU bacteria was applied to each plate of supplemented Brucella blood agar (BBL Microbiology Systems, Cockeysville, Md.) using a Steers replicator. The plates were incubated in an anaerobic chamber for 48 h at 35 C. For daptomycin susceptibility assays, the broth microdilution method using Brucella broth with hemin (5 μg/ml), vitamin K1 (1 μg/ml), lysed horse blood (5%), and calcium (50 μg/ml) was used (6). The 12 antimicrobial agents used for susceptibility testing were obtained from their corresponding manufacturers: fidaxomicin (Optimer Pharmaceuticals Inc. San Diego, CA, USA); Rifaximin, Vancomycin, and metronidazole (Sigma, St Louis, MO, USA); ciprofloxacin and moxifloxacin (Bayer Co., West Haven, CT, USA); levofloxacin (Daiichi Pharmaceuticals, Tokyo, Japan); gemifloxacin (LG Chem Investments, Seoul, Korea); nemonoxacin (TaiGen Biotechnology, Co. Ltd, Taipei, Taiwan); daptomycin (Cubist Pharmaceuticals, Lexington, MA, USA); and tigecycline (Pfizer Inc., New York, NY, USA) 5

106 107 108 109 110 111 112 113 114 115 116 117 118 The MIC was defined as the lowest concentration of each antimicrobial agent that inhibited the growth of the tested isolate. C. difficile ATCC 700057 and Bacteroides fragilis ATCC 25285 were used for quality control for each run of susceptibility testing. The MIC interpretive breakpoints for metronidazole, clindamycin, and moxifloxacin followed the guidelines recommended by the CLSI (6), and breakpoints for vancomycin (susceptible, MICs 2 μg/ml; and resistant, MICs >2 μg/ml) were those recommended by The European Committee on Antimicrobial Susceptibility Testing (EUCAST) (13) (Table 1). Breakpoints are not established for rifaximin and fidaxomicin. Genotyping and sequencing. Presence of tcda, tcdb, cdta, and cdtb genes were determined by multiplex PCR as described (13). Moxifloxacin-nonsusceptible isolates (moxifloxacin MICs of 4 μg/ml) were subjected to partial sequencing of gyra and gyrb genes after PCR amplification of 390 bp fragments of each (13) 6

119 RESULTS 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 Antimicrobial susceptibilities. Susceptibilities to 16 antimicrobial agents, including 4 used clinically to treat CDI (metronidazole, vancomycin, rifaximin, and fidaxomicin), were determined for 403 clinical isolates of C. difficile. Susceptibilities of the test strain C. difficile ATCC 700057 to fidaxomicin, rifaximin, vancomycin, and metronidazole were with the CLSI standard ranges, and susceptibility of B. fragilis ATCC 25285 to metronidazole was also within the established range. Table 1 presents the ranges of MIC values of individual isolates and MIC 50 and MIC 90 values for each agent. Distribution of isolates according to susceptibility is presented for four antibiotics for which MIC breakpoints have been established. Only 13.6% of isolates were fully susceptible to clindamycin, and the MIC 90 value was >256 μg/ml. Among the fluoroquinolones, susceptibility was lowest for levofloxacin (MIC 90, 128 μg/ml), followed by ciprofloxacin (MIC 90, 64 μg/ml), gemifloxacin (MIC 90, 2 μg/ml), and moxifloxacin (MIC 90, 16 μg/ml); 80% of isolates were fully susceptible to moxifloxacin. Isolates were more susceptible to the nonfluorinated quinolone, nemonoxacin (MIC 90, 8 μg/ml; range 0.25- >32 μg/ml). The majority of isolates were inhibited by tigecycline (MIC 90, 0.06 μg/ml) and daptomycin (MIC 90, 1 μg/ml), although the range of MIC values was 0.06-8 μg/ml for daptomycin. Distribution of isolates by MIC value is plotted in Figure 1A for the quinolones. Susceptibility can be ranked in order as nemonoxacin (most susceptible), moxifloxacin/gemifloxacin, levofloxacin, and ciprofloxacin (least susceptible). Two isolates (0.5%) were resistant to vancomycin (MICs, 4 μg/ml) by EUCAST criteria and none were resistant to metronidazole. Ninety percent of isolates were susceptible to metronidazole at 0.5 μg/ml (range, 0.03-4 μg/ml) and 90% were inhibited by vancomycin at 1 7

141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 μg/ml (range, 0.06-4 μg/ml). Fidaxomicin exhibited potent in vitro activities against these isolates; the range of MIC values was 0.015 to 0.5 μg/ml and 90% of isolates were inhibited at 0.25 μg/ml. In contrast, there was a wide range of MICs to rifaximin ( 0.015- >128 μg/ml; MIC 90 >128 μg/ml): 352 (87.3%) isolates with MICs of 0.25 μg/ml, four (1.0%) with MICs of 4 μg/ml, three (0.7%) with MICs of 128 μg/ml, and 44 (10.9%) with MICs of > 128 μg/ml (Figure 1B). Genotypes and antimicrobial susceptibilities. Of the 403 isolates, 57.8% (233/403) were potentially toxigenic by genotype, carrying both tcda and tcdb genes. Of those, 16.7% (39/233) also possessed the genes for the binary toxin, cdta and cdtb; this represents 9.6% (39/403) of all isolates. The remaining 42.2% (170/403) of isolates did not carry the genes encoding toxins A and B and are presumably not toxigenic. There was no clear pattern linking antibiotic susceptibility to genotype according to the toxin genes, with the exception that many tcda+b+ cdta B isolates were resistant to rifaximin, and the MIC 90 value was >128 μg/ml. Susceptibility to fidaxomicin was identical across all three genotypes, and susceptibilities to vancomycin and metronidazole differed by no more than two-fold between genotypes. The 39 isolates carrying the binary toxin genes tended to be more susceptible to clindamycin (MIC 90, 32 μg/ml) than those without cdta and cdtb genes (MIC 90 >256 μg/ml). There was no more than a two-fold difference in MIC 90 values between genotypes for the remaining eight agents. For those agents with resistance breakpoints defined, results are presented by distribution of isolates among susceptible, intermediate, and resistant classes according to genotype. There was no pattern linking resistance to genotype around the toxins A and B and binary toxin genes, although neither of the two vancomycin-resistant isolates carried the binary toxin genes. Isolates 8

163 164 165 with reduced susceptibility to moxifloxacin (MIC 4 μg/ml), as well as the other fluoroquinolones, were found in all genotype classes: tcda + B + cdta + B + (10/39, 25.6%); tcda + B + cdta B (37/194, 19.1%); and tcda B cdta B (34/170, 20.0%). 166 167 168 169 170 171 172 173 174 175 176 Gyrase mutations. Of the 403 isolates, 81 (20.1%) had reduced susceptibility to moxifloxacin (MIC 4 μg/ml) and 72 (88.9%) of those were fully resistant (MIC 8 μg/ml). All but 8 isolates with reduced susceptibility to moxifloxacin harbored amino acid substitutions in gyra alone (52 isolates), gyrb alone (16 isolates), or both (5 isolates). One had multiple gyra mutations (at Asp81, Arg90, Asp103, and Glu123) and a single gyrb substitution (at Asp426), and another had 6 gyrb amino acid substitutions as well as a Thr82 to Ile substitution in gyra. The most common substitution in gyra was Thr82 to Ile (52/57 isolates with gyra mutations) and the most common gyrb substitution was Asp426 to Asn (9/21 isolates with gyrb mutations). There was a high level of cross-resistance to ciprofloxacin and levofloxacin, but some isolates remained susceptible to gemifloxacin or nemonoxacin at 0.5 μg/ml. 9

177 DISCUSSION 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 We characterized antibiotic susceptibility patterns of 403 clinical isolates of C. difficile in Taiwan collected over a six-year period (2005-2010); around 58% of the isolates were toxigenic by genotype. We found all 403 isolates to be fully susceptible to metronidazole with MICs 4 μg/ml. All but two isolates were susceptible to vancomycin, and only one vancomycin-resistant isolate was a toxigenic strain; that particular isolate carried toxin A, toxin B, and binary toxin genes and had a MIC for vancomycin of 4 μg/ml, still orders of magnitude below fecal levels of vancomycin achieved during treatment (17). No vancomycin- or metronidazole-resistant clones were found among 100 clinical isolates from South Korea during 2006-2008 (23). Among 112 clinical isolates cultured in China late 2008 to early 2009, none had reduced susceptibility to metronidazole, but two isolates had a vancomycin MIC of 4 μg/ml (22). Our earlier study found also that all 113 isolates of C. difficile collected in Taiwan during 2001-2009 were susceptible to metronidazole, but, as in this study, some had reduced susceptibility to vancomycin (MIC, 4 μg/ml). In this study, fidaxomicin, that has recently approved by the US Federal Drug Administration and the European Medicines Agency for the treatment of CDI, had potent in vitro activity against all the isolates tested (18). The MIC 50 (0.12 μg/ml) and MIC 90 (0.25 μg/ml) values for fidaxomicin were identical to those found for 716 isolates from patients at enrollment in clinical trials of fidaxomicin in North America and Europe (16). Another RNA polymerase inhibitor, rifaximin, has been used against CDI following standard vancomycin therapy. We found that 10.9% of isolates in this study were not effectively inhibited by rifaximin at 128 μg/ml. All rifaximin-resistant isolates lacked the binary toxin genes but some contained toxin A 10

199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 and toxin B genes and some were non-toxigenic. In the same study cited for fidaxomicin, rifaximin-resistant C. difficile clones were isolated in United States, Germany, and Italy, but not in the United Kingdom, Belgium, France, Spain, Sweden, or Canada. That study and ours found no evidence of cross resistance to fidaxomicin; this is not unexpected since the two transcriptional inhibitors interact with different regions of RNA polymerase (37). One-fifth of the isolates had reduced susceptibility to moxifloxacin (MIC 4 μg/ml), with cross resistance to ciprofloxacin and levofloxacin. Some moxifloxacin-nonsusceptible isolates were susceptible to gemifloxacin and nemonoxacin. There was no correlation between fluoroquinolone resistance and the presence of the binary toxin gene, two identifying characteristics of the hypervirulent 027 strain. The first identified and most common mutation associated with fluoroquinolone resistance in C. difficile (1, 8, 11, 35, 36, 38) was the single most common gyrase mutation among these fluoroquinolone reduced susceptibility isolates, gyra Thr82 to Ile, present in 52 isolates (64%). One other isolate had a Thr82 to Ala mutation, not previously reported, although a Thr82 to Val mutation was reported in an isolate from China (22). Some isolates with the gyra Thr82 to Ile substitution alone had low to intermediate resistance (MICs of 4-8 μg/ml) to moxifloxacin in contrast to other studies that found this substitution to be associated only with high level resistance to moxifloxacin (MICs of 16 μg/ml) among isolates from France and Canada (10, 38). The single most common gyrb substitution was Asp426 to Asn in nine isolates, which as a single substitution was associated with moxifloxacin MICs from 4-16 μg/ml. Asp426 to Asn or Val substitutions have been associated with reduced fluoroquinolone susceptibility in several studies (10, 12, 21, 35, 38). 11

220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 We also found previously unreported amino acid substitutions in the gyrb gene, two of which apparently can independently confer resistance to moxifloxacin and other fluoroquinolones: Ser416 to Ala and Glu466 to Lys were each found as the only gyrase amino acid substitution in one isolate each with MIC of 8 μg/ml. Ser416 to Ala was also identified in two other isolates which carried the Thr82 to Ile (gyra) mutation as well. Six amino substitutions in GyrB in one fluoroquinolone-resistant isolate may be silent since they were accompanied by the GyrA Thr82 to Ile substitution. Site-directed mutagenesis would be required to determine whether any of these substitutions independently or together reduce susceptibility to moxifloxacin; none have been reported in fluoroquinolone-resistant C. difficile isolates before. Of the 81 moxifloxacin-nonsusceptible isolates, eight had no mutations in the regions of gyra and gyrb established as being important for susceptibility to quinolones; three of those were resistant to moxifloxacin at 32 μg/ml while the remaining 5 were intermediate in susceptibility to moxifloxacin (MICs, 4 μg/ml). We cannot rule out that other mutations outside these regions exist in the gyrase genes. Other mechanisms of quinoline resistance have been identified; a pentapeptide repeat protein encoded by qnra acts in trans to protect DNA gyrase from quinolone activity (19). Qnr genes are found on resistance plasmids as well as chromosomally in some gram-positive species, including a toxigenic laboratory strain of C. difficile, ATCC 9689 (19, 34). This study describes the diversity of toxigenic and nontoxigenic strains of C. difficile found in the healthcare settings in Taiwan. There is no evidence of increasing resistance to the antibiotics commonly used to treat CDI, metronidazole and vancomycin. In addition, 12

241 242 fidaxomicin exhibited potent in vitro activity against all isolates while there was concerning resistance to another transcription inhibitor, rifaximin. 13

243 ACKNOWLEDGMENTS 244 245 This study was supported in part by grants from Optimer Biotechnology, Inc., Taiwan, ROC. We thank Sharon Dana, PhD for assisting with preparation of the manuscript. 14

246 REFERENCES 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 1. Ackermann, G., Y. J. Tang, R. Kueper, P. Heisig, A. C. Rodloff, J. Silva, Jr., and S. H. Cohen. 2001. Resistance to moxifloxacin in toxigenic Clostridium difficile isolates is associated with mutations in gyra. Antimicrob. Agents Chemother. 45:2348-2353. 2. Al-Nassir, W. N., A. K. Sethi, Y. Li, M. J. Pultz, M. M. Riggs, and C. J. Donskey. 2008. Both oral metronidazole and oral vancomycin promote persistent overgrowth of vancomycin-resistant enterococci during treatment of Clostridium difficile-associated disease. Antimicrob. Agents Chemother. 52:2403-2406. 3. Baines, S. D., R. O'Connor, J. Freeman, W. N. Fawley, C. Harmanus, P. Mastrantonio, E. J. Kuijper, and M. H. Wilcox. 2008. Emergence of reduced susceptibility to metronidazole in Clostridium difficile. J. Antimicrob. Chemother. 62:1046-1052. 4. Bauer, M. P., E. J. Kuijper, and J. T. van Dissel. 2009. European Society of Clinical Microbiology and Infectious Diseases (ESCMID): treatment guidance document for Clostridium difficile infection (CDI). Clin. Microbiol. Infect. 15:1067-1079. 5. Chung, C. H., C. J. Wu, H. C. Lee, J. J. Yan, C. M. Chang, N. Y. Lee, P. L. Chen, C. C. Lee, Y. P. Hung, and W. C. Ko. 2010. Clostridium difficile infection at a medical center in southern Taiwan: incidence, clinical features and prognosis. J. Microbiol. Immunol. Infect. 43:119-125. 6. Clinical and Laboratory Standards Institute. 2007. Methods for antimicrobial susceptibility testing of anaerobic bacteria. Approved standard M11-A7, 7th ed. Clinical and Laboratory Standards Institute, Wayne, PA. 15

268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 7. Cohen, S. H., D. N. Gerding, S. Johnson, C. P. Kelly, V. G. Loo, L. C. McDonald, J. Pepin, and M. H. Wilcox. 2010. Clinical practice guidelines for Clostridium difficile infection in adults: 2010 update by the society for healthcare epidemiology of America (SHEA) and the infectious diseases society of America (IDSA). Infect. Control Hosp. Epidemiol. 31:431-455. 8. Deneve, C., S. Bouttier, B. Dupuy, F. Barbut, A. Collignon, and C. Janoir. 2009. Effects of subinhibitory concentrations of antibiotics on colonization factor expression by moxifloxacin-susceptible and moxifloxacin-resistant Clostridium difficile strains. Antimicrob. Agents Chemother. 53:5155-5162. 9. Dingle, K. E., D. Griffiths, X. Didelot, J. Evans, A. Vaughan, M. Kachrimanidou, N. Stoesser, K. A. Jolley, T. Golubchik, R. M. Harding, T. E. Peto, W. Fawley, A. S. Walker, M. Wilcox, and D. W. Crook. 2011. Clinical Clostridium difficile: clonality and pathogenicity locus diversity. PLoS One 6:e19993. 10. Dridi, L., J. Tankovic, B. Burghoffer, F. Barbut, and J. C. Petit. 2002. gyra and gyrb mutations are implicated in cross-resistance to Ciprofloxacin and moxifloxacin in Clostridium difficile. Antimicrob. Agents Chemother. 46:3418-3421. 11. Drudy, D., L. Kyne, R. O'Mahony, and S. Fanning. 2007. gyra mutations in fluoroquinolone-resistant Clostridium difficile PCR-027. Emerg. Infect. Dis. 13:504-505. 12. Drudy, D., T. Quinn, R. O'Mahony, L. Kyne, P. O'Gaora, and S. Fanning. 2006. High-level resistance to moxifloxacin and gatifloxacin associated with a novel mutation in gyrb in toxin-a-negative, toxin-b-positive Clostridium difficile. J. Antimicrob. Chemother. 58:1264-1267. 16

290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 13. European Committee on Antimicrobial Susceptibility Testing. http://www.eucast.org/fileadmin/src/media/pdfs/eucast_files/disk_test_documents/e UCAST_breakpoints_v_2.0_120101.pdf. 14. Garey, K. W., S. S. Ghantoji, D. N. Shah, M. Habib, V. Arora, Z. D. Jiang, and H. L. DuPont. 2011. A randomized, double-blind, placebo-controlled pilot study to assess the ability of rifaximin to prevent recurrent diarrhoea in patients with Clostridium difficile infection. J. Antimicrob. Chemother. 66:2850-2855. 15. Garey, K. W., Z. D. Jiang, A. Bellard, and H. L. Dupont. 2009. Rifaximin in treatment of recurrent Clostridium difficile-associated diarrhea: an uncontrolled pilot study. J. Clin. Gastroenterol. 43:91-93. 16. Goldstein, E. J., D. M. Citron, P. Sears, F. Babakhani, S. P. Sambol, and D. N. Gerding. 2011. Comparative susceptibilities to fidaxomicin (OPT-80) of isolates collected at baseline, recurrence, and failure from patients in two phase III trials of fidaxomicin against Clostridium difficile infection. Antimicrob. Agents Chemother. 55:5194-5199. 17. Gonzales, M., J. Pepin, E. H. Frost, J. C. Carrier, S. Sirard, L. C. Fortier, and L. Valiquette. 2010. Faecal pharmacokinetics of orally administered vancomycin in patients with suspected Clostridium difficile infection. BMC Infect. Dis. 10:363. 18. Hardesty, J. S., and P. Juang. 2011. Fidaxomicin: a macrocyclic antibiotic for the treatment of Clostridium difficile infection. Pharmacotherapy 31:877-886. 19. Hernandez, A., M. B. Sanchez, and J. L. Martinez. 2011. Quinolone resistance: much more than predicted. Front. Microbiol. 2:22. 17

312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 20. Hsu, M. S., J. T. Wang, W. K. Huang, Y. C. Liu, and S. C. Chang. 2006. Prevalence and clinical features of Clostridium difficile-associated diarrhea in a tertiary hospital in northern Taiwan. J. Microbiol. Immunol. Infect. 39:242-248. 21. Huang, H., A. Weintraub, H. Fang, and C. E. Nord. 2009. Antimicrobial resistance in Clostridium difficile. Int. J. Antimicrob. Agents 34:516-522. 22. Huang, H., A. Weintraub, H. Fang, S. Wu, Y. Zhang, and C. E. Nord. 2010. Antimicrobial susceptibility and heteroresistance in Chinese Clostridium difficile strains. Anaerobe 16:633-645. 23. Kim, H., S. H. Jeong, K. H. Roh, S. G. Hong, J. W. Kim, M. G. Shin, M. N. Kim, H. B. Shin, Y. Uh, H. Lee, and K. Lee. 2010. Investigation of toxin gene diversity, molecular epidemiology, and antimicrobial resistance of Clostridium difficile isolated from 12 hospitals in South Korea. Korean J. Lab. Med. 30:491-497. 24. Kuijper, E. J., B. Coignard, J. S. Brazier, C. Suetens, D. Drudy, C. Wiuff, H. Pituch, P. Reichert, F. Schneider, A. F. Widmer, K. E. Olsen, F. Allerberger, D. W. Notermans, F. Barbut, M. Delmee, M. Wilcox, A. Pearson, B. C. Patel, D. J. Brown, R. Frei, T. Akerlund, I. R. Poxton, and P. Tull. 2007. Update of Clostridium difficileassociated disease due to PCR ribotype 027 in Europe. Euro. Surveill. 12:E1-2. 25. Lee, Y. C., J. T. Wang, A. C. Chen, W. H. Sheng, S. C. Chang, and Y. C. Chen. 2012. Changing incidence and clinical manifestations of Clostridium difficile-associated diarrhea detected by combination of glutamate dehydrogenase and toxin assay in Northern Taiwan. J. Microbiol. Immunol. Infect. 2011 26. Lin, Y. C., Y. T. Huang, P. J. Tsai, T. F. Lee, N. Y. Lee, C. H. Liao, S. Y. Lin, W. C. Ko, and P. R. Hsueh. 2011. Antimicrobial susceptibilities and molecular epidemiology 18

335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 of clinical isolates of Clostridium difficile in Taiwan. Antimicrob. Agents Chemother. 55:1701170-5. 27. McDonald, L. C., G. E. Killgore, A. Thompson, R. C. Owens, Jr., S. V. Kazakova, S. P. Sambol, S. Johnson, and D. N. Gerding. 2005. An epidemic, toxin gene-variant strain of Clostridium difficile. N. Engl. J. Med. 353:2433-2441. 28. Miller, B. A., L. F. Chen, D. J. Sexton, and D. J. Anderson. 2011. Comparison of the burdens of hospital-onset, healthcare facility-associated Clostridium difficile Infection and of healthcare-associated infection due to methicillin-resistant Staphylococcus aureus in community hospitals. Infect. Control Hosp. Epidemiol. 32:387-390. 29. Miller, M., D. Gravel, M. Mulvey, G. Taylor, D. Boyd, A. Simor, M. Gardam, A. McGeer, J. Hutchinson, D. Moore, and S. Kelly. 2010. Health care-associated Clostridium difficile infection in Canada: patient age and infecting strain type are highly predictive of severe outcome and mortality. Clin. Infect. Dis. 50:194-201. 30. Musher, D. M., S. Aslam, N. Logan, S. Nallacheru, I. Bhaila, F. Borchert, and R. J. Hamill. 2005. Relatively poor outcome after treatment of Clostridium difficile colitis with metronidazole. Clin. Infect. Dis. 40:1586-1590. 31. O'Connor, J. R., S. Johnson, and D. N. Gerding. 2009. Clostridium difficile infection caused by the epidemic BI/NAP1/027 strain. Gastroenterology 136:1913-1924. 32. Pepin, J., L. Valiquette, and B. Cossette. 2005. Mortality attributable to nosocomial Clostridium difficile-associated disease during an epidemic caused by a hypervirulent strain in Quebec. CMAJ 173:1037-1042. 33. Pituch, H., W. van Leeuwen, K. Maquelin, D. Wultańska, P. Obuch-Woszczatyński, G. Nurzyńska, H. Kato, M. Reijans, F. Meisel-Mikołajczyk, M. Łuczak, and A. van 19

358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 Belkum. 2007. Toxin profiles and resistances to macrolides and newer fluoroquinolones as epidemicity determinants of clinical isolates of Clostridium difficile from Warsaw, Poland. J. Clin. Microbiol. 45:1607-1610. 34. Rodriguez-Martinez, J. M., C. Velasco, A. Briales, I. Garcia, M. C. Conejo, and A. Pascual. 2008. Qnr-like pentapeptide repeat proteins in gram-positive bacteria. J. Antimicrob. Chemother. 61:1240-1243. 35. Spigaglia, P., F. Barbanti, A. M. Dionisi, and P. Mastrantonio. 2010. Clostridium difficile isolates resistant to fluoroquinolones in Italy: emergence of PCR ribotype 018. J. Clin. Microbiol. 48:2892-2896. 36. Spigaglia, P., F. Barbanti, P. Mastrantonio, J. S. Brazier, F. Barbut, M. Delmee, E. Kuijper, and I. R. Poxton. 2008. Fluoroquinolone resistance in Clostridium difficile isolates from a prospective study of C. difficile infections in Europe. J. Med. Microbiol.57:784-789. 37. Srivastava, A., M. Talaue, S. Liu, D. Degen, R. Y. Ebright, E. Sineva, A. Chakraborty, S. Y. Druzhinin, S. Chatterjee, J. Mukhopadhyay, Y. W. Ebright, A. Zozula, J. Shen, S. Sengupta, R. R. Niedfeldt, C. Xin, T. Kaneko, H. Irschik, R. Jansen, S. Donadio, N. Connell, and R. H. Ebright. 2011. New target for inhibition of bacterial RNA polymerase: 'switch region'. Curr. Opin. Microbiol. 14:532-543. 38. Walkty, A., D. A. Boyd, D. Gravel, J. Hutchinson, A. McGeer, D. Moore, A. Simor, K. Suh, G. Taylor, M. Miller, and M. R. Mulvey. 2010. Molecular characterization of moxifloxacin resistance from Canadian Clostridium difficile clinical isolates. Diagn. Microbiol. Infect. Dis. 66:419-424. 20

380 381 382 383 384 385 386 387 388 39. Warny, M., J. Pepin, A. Fang, G. Killgore, A. Thompson, J. Brazier, E. Frost, and L. C. McDonald. 2005. Toxin production by an emerging strain of Clostridium difficile associated with outbreaks of severe disease in North America and Europe. Lancet 366:1079-1084. 40. Zar, F. A., S. R. Bakkanagari, K. M. Moorthi, and M. B. Davis. 2007. A comparison of vancomycin and metronidazole for the treatment of Clostridium difficile-associated diarrhea, stratified by disease severity. Clin. Infect. Dis. 45:302-307. Downloaded from http://aac.asm.org/ on November 24, 2018 by guest 21

389 390 391 TABLE 1. In vitro susceptibilities of 403 isolates of C. difficile to fidaxomicin, rifaximin, and 10 other antimicrobial agents MIC (μg/ml) a No. (%) of isolates Agent Range 50% 90% Susceptible Resistant Fidaxomicin 0.015-0.5 0.12 0.25 - - Rifaximin 0.015->128 0.015 >128 - - Metronidazole b 0.03-4 0.5 0.5 403 (100) 0 (0) Vancomycin c 0.06-4 0.5 1 401 (99.5) 2 (0.5) Clindamycin b 0.06->256 8 >256 55 (13.6) 296 (73.5) Ciprofloxacin 0.5-128 16 64 - - Moxifloxacin b 0.06-32 2 16 322 (79.9) 72 (17.9) Levofloxacin 1->128 4 128 - - Gemifloxacin 0.25->32 2 32 - - Nemonoxacin 0.25->32 1 8 - - Tigecycline 0.03-1 0.06 0.06 - - 22

Daptomycin 0.06-8 1 1 - - a MICs were determined by the agar dilution method with the exception of daptomycin, for which the broth microdilution method was used. 392 393 b MIC breakpoints applied were those recommended for anaerobes by the Clinical and Laboratory Standards Institute (CLSI-2007, M11-A7) (6). For metronidazole, susceptible, 8 μg/ml; resistant, 32 μg/ml. For clindamycin, susceptible, 2 μg/ml; resistant, 8 μg/ml. For moxifloxacin, susceptible, 2 μg/ml; resistant, 8 μg/ml. c For vancomycin there are no CLSI-recommended MIC breakpoints. Breakpoints are those recommended by The European Committee on Antimicrobial Susceptibility Testing (EUCAST) (susceptible, 2 μg/ml; resistant, >2 μg/ml) (13). The two isolates resistant to vancomycin both had vancomycin MICs of 4 μg/ml. 23

394 395 TABLE 2. Pathogenicity locus and binary toxin genotypes and in vitro susceptibilities of C. difficile isolates to fidaxomicin, rifaximin, and other antimicrobial agents MIC (μg/ml) tcda + tcdb + cdta + cdtb + tcda + tcdb + cdta cdtb tcda tcdb cdta cdtb (n=39) (n=194) (n=170) Agent Range 50% 90% Range 50% 90% Range 50% 90% Fidaxomicin 0.015-0.5 0.12 0.25 0.015-0.5 0.12 0.25 0.015-0.5 0.12 0.25 Rifaximin 0.015-2 0.015 0.015 0.015->128 0.015 >128 0.015->128 0.015 0.06 Metronidazole b 0.12-1 0.25 1 0.03-1 0.5 0.5 0.06-4 0.5 1 Vancomycin c 0.25-2 0.5 0.5 0.06-4 0.5 1 0.25-4 0.5 1 Clindamycin b 0.12->256 8 32 0.5->256 16 >256 0.06->256 8 >256 Ciprofloxacin 8-128 8 64 4-128 16 64 0.5-128 16 64 24

396 397 Moxifloxacin b 1-32 2 16 0.06-32 2 16 0.5-32 2 16 Levofloxacin 2->128 4 >128 1->128 4 128 1->128 4 128 Gemifloxacin 1->32 2 >32 0.5->32 2 16 0.25->32 2 32 Nemonoxacin 0.25-16 1 8 0.25->32 1 8 0.25->32 1 8 Tigecycline 0.03-0.12 0.06 0.12 0.03-0.5 0.06 0.06 0.03-1 0.06 0.06 Daptomycin 0.25-4 1 2 0.06-4 0.5 1 0.06-8 0.5 1 MICs were determined by the agar dilution method with the exception of daptomycin, for which the broth microdilution method was used. 25

398 399 TABLE 3. Susceptibility distribution of 403 clinical isolates of C. difficile by genotypes to four agents with MIC interpretive breakpoints by the Clinical and Laboratory Standards Institute (6) No. (%) of isolates for each genotype tcda + tcdb + cdta + cdtb + tcda + tcdb + cdta cdtb tcda tcdb cdta cdtb (n=39) (n=194) (n=170) Agent Susceptible Resistant Susceptible Resistant Susceptible Resistant Metronidazole a 39 (100) 0 (0) 194 (100) 0 (0) 170 (100) 0 (0) Vancomycin b 39 (100) 0 (0) 193 (99) 1 (1) 169 (99) 1 (1) Clindamycin a 8 (21) 24 (62) 14 (7) 151 (78) 33 (19) 121 (71) Moxifloxacin a 29 (74) 10 (26) 157 (81) 33 (17) 136 (80) 29 (17) a MIC breakpoints applied were those recommended for anaerobes by the Clinical and Laboratory Standards Institute (CLSI-2007, M11-A7) (6). b For vancomycin there are no CLSI-recommended MIC breakpoints. Breakpoints are those recommended by The European Committee on Antimicrobial Susceptibility Testing (EUCAST) (13). 26

400 401 TABLE 4. Minimum inhibitory concentrations (MICs) of quinolones and substitutions in GyrA and GyrB for 81 isolates of C. difficile with reduced susceptibility to moxifloxacin (MIC 4 μg/ml) MIC (μg/ml) a Amino acid substitutions b No. of isolates No. of isolates with GyrA GyrB with indicated Moxi Cipro Levo Gemi Nemo indicated amino MIC values acid substitutions 9 4 8-32 4->64 2->32 0.5-8 2 Thr82 to Ile NF 1 Asp71 to Gly NF 1 Thr82 to Ala NF 5 NF NF 19 8 16-128 32->128 0.5-32 4->32 2 Thr82 to Ile NF 1 Asp71 to Val NF 1 Asp81 to Asn NF 7 NF Asp426 to Asn 27

3 NF Asp426 to Val 1 NF Glu466 to Lys 1 NF Ser416 to Ala 1 Thr82 to Ile Asp426 to Val 1 Thr82 to Ile Ser416 to Ala 1 Asp81 to Asn Asp426 to Val Arg90 to Lys Asp103 to Asn Glu123 to Lys 33 16 16-128 4->128 0.5->32 0.5-16 27 Thr82 to Ile NF 2 NF Asp426 to Asn 1 NF Ser416 to Ala 1 NF Arg377 to Gly 1 Thr82 to Ile Ser416 to Ala 1 Thr82 to Ile Arg389 to Pro 28

402 403 Glu399 to Lys Asp409 to Asn Val423 to Phe Arg457 to Thr Asp465 to Tyr 20 32 16-128 64->128 2->32 0.5-16 17 Thr82 to Ile NF 3 NF NF a Moxi, moxifloxacin; Cipro, ciprofloxacin; Levo, levofloxacin; Gemi, gemifloxacin; Nemo, nemonoxacin. b NF, Amino acid substitutions in gyra or gyrb were not found. 29

404 FIGURE LEGEND 405 406 407 408 FIG. 1. Distribution of minimum inhibitory concentrations (MICs) among clinical isolates of C. difficile to five quinolones (A) and fidaxomicin and rifaximin (B). 30

409 Downloaded from http://aac.asm.org/ on November 24, 2018 by guest 31

% of Iso olates (A) 100 Ciprofloxacin Levofloxacin Moxifloxacin 80 Gemifloxacin Nemonoxacin 60 40 20 00 0.015 0.06 0.25 1 0.03 0.12 0.5 4 16 64 256 2 8 32 128 MIC (μg/ml)

% of Isola ates (B) 100 80 60 40 20 00 0.015 0.06 0.25 1 0.03 0.12 0.5 MIC (μg/ml) Fidaxomicin Rifaximin 4 16 64 256 2 8 32 128