Keywords: CB-183,315; Clostridium difficile; susceptibility; intestinal anaerobes

Similar documents
ANTI-ANAEROBIC ACTIVITIES OF SULOPENEM COMPARED TO SIX OTHER. Departments of Pathology, Hershey Medical Center, Hershey, PA 17033

In Vitro Activities of OPT-80 and Comparator Drugs against Intestinal Bacteria

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

SESSION XVI NEW ANTIBIOTICS

R. M. Alden Research Laboratory, Santa Monica, California 90404, 1 and David Geffen School of Medicine at UCLA, Los Angeles, California

on February 12, 2018 by guest

Antimicrobial Susceptibility Patterns of Anaerobic Bacterial Clinical Isolates From 2014 to 2016, Including Recently Named or Renamed Species

Multicenter Study of Antimicrobial Susceptibility of Anaerobic Bacteria in Korea in 2012

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

Piperacillin-Tazobactam, and Cefoxitin

Title: Effect of fidaxomicin versus vancomycin on susceptibility to intestinal

Patterns of Susceptibility to Fluoroquinolones Among Anaerobic Bacterial Isolates in the United States

Third Belgian multicentre survey of antibiotic susceptibility of anaerobic bacteria

Present Status of Therapy for Anaerobic Infections

As presented at 112th General Meeting of the American Society for Microbiology, Session C-173 June 16 19, 2012, San Francisco, California

Should we test Clostridium difficile for antimicrobial resistance? by author

Anaerobic bacteria in 118 patients with deepspace head and neck infections from the University Hospital of Maxillofacial Surgery, Sofia, Bulgaria

Labeled Uses: Acute Skin & Skin Structure Infections caused by susceptible bacteria 1,2,4

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

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

Effect of dalbavancin on the normal intestinal microflora

Moxifloxacin resistance is prevalent among Bacteroides and Prevotella species in Greece

ECOLOGICAL IMPACT OF NARROW SPECTRUM ANTIMICROBIAL AGENTS COMPARED TO BROAD SPECTRUM AGENTS ON THE HUMAN INTESTINAL MICROFLORA CARL ERIK NORD

Reply to Fabre et. al

INFECTIOUS DISEASES DIAGNOSTIC LABORATORY NEWSLETTER

Surveillance of susceptibility patterns in 1297 European and US anaerobic and capnophilic isolates to co-amoxiclav and five other antimicrobial agents

Tel: Fax:

Ciprofloxacin, Enoxacin, and Ofloxacin against Aerobic and

This article appeared in a journal published by Elsevier. The attached copy is furnished to the author for internal non-commercial research and

Aberdeen Hospital. Antibiotic Susceptibility Patterns For Commonly Isolated Organisms For 2015

Susceptibility Testing of Anaerobic Bacteria: Evaluation of the Redesigned (Version 96) biomérieux ATB ANA Device

Antimicrobial Resistance in Human Oral and Intestinal Anaerobic Microfloras

Classification of Bacteria

Clostridium difficile infection: The Present and the Future

ACCEPTED. Anaerobe Reference Laboratory, Department of Bacterial and Inflammatory Diseases, National

Resistance pattern of anaerobic bacteria isolated in a general hospital during a two-year period

Intra-abdominal infections: review of the bacteriology, antimicrobial susceptibility and the role of ertapenem in their therapy

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

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

Effect of Lactobacillus F19 on the emergence of antibioticresistant microorganisms in the intestinal microflora

Taking Action to Prevent and Manage Multidrug-resistant Organisms and C. difficile in the Nursing Home: Part 1 Reviewing the organisms

MICHAEL J. RYBAK,* ELLIE HERSHBERGER, TABITHA MOLDOVAN, AND RICHARD G. GRUCZ

Specificity (target gene) Primer name Sequence Product length[bp] GGATTAGATACCCTGGTAGTC TACCTTGTTACGACTT

Evaluation of a computerized antimicrobial susceptibility system with bacteria isolated from animals

Help with moving disc diffusion methods from BSAC to EUCAST. Media BSAC EUCAST

Chapter Anaerobic infections (individual fields): intraperitoneal infections (acute peritonitis, hepatobiliary infections, etc.

The prevalence of antibiotic resistance in anaerobic bacteria isolated from patients with skin infections Research Article

Ecological impact of the des-f(6)-quinolone, BMS , on the normal intestinal microflora C. E. Nord 1, D. A. Gajjar 2 and D. M.

NON-SPORING ANAEROBES

Clindamycin Palmitate Hydrochloride for Oral Solution, USP (Pediatric)

ETX0282, a Novel Oral Agent Against Multidrug-Resistant Enterobacteriaceae

2015 Antibiogram. Red Deer Regional Hospital. Central Zone. Alberta Health Services

Epidemiology and Antimicrobial Susceptibility of Anaerobic Bloodstream Infections: A 10 Years Study

against Clinical Isolates of Gram-Positive Bacteria

Title: N-Acetylcysteine (NAC) Mediated Modulation of Bacterial Antibiotic

In Vitro Antibacterial Properties of Pexiganan, an Analog of Magainin

Test Method Modified Association of Analytical Communities Test Method Modified Germicidal Spray Products as Disinfectants

2016 Antibiogram. Central Zone. Alberta Health Services. including. Red Deer Regional Hospital. St. Mary s Hospital, Camrose

Learning Objectives 6/1/18

Intra-abdominal anaerobic infections. Diagnostics and therapy

2017 Antibiogram. Central Zone. Alberta Health Services. including. Red Deer Regional Hospital. St. Mary s Hospital, Camrose

Effect of Antibiotic Treatment on Establishment and Elimination of Intestinal Colonization by KPC-Producing Klebsiella pneumoniae in Mice

Secondary bacterial infections complicating skin lesions

An evaluation of the susceptibility patterns of Gram-negative organisms isolated in cancer centres with aminoglycoside usage

PDF hosted at the Radboud Repository of the Radboud University Nijmegen

Susceptibility of Respiratory Tract Anaerobes to Orally Administered Penicillins and Cephalosporins

Anaerobic and microaerophilic gram-positive cocci Peptococcus species, Peptostreptococcus species, Microaerophilic streptococci

1/30/ Division of Disease Control and Health Protection. Division of Disease Control and Health Protection

Alfonso Torress-Cook, Dr.P.H. Director of Epidemiology/Patient Safety Pacific Hospital of Long Beach

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

Fluoroquinolones and Anaerobes

Lessons Learned from the Anaerobe Survey: Historical Perspective and Review of the Most Recent Data ( )

ESCMID Online Lecture Library. by author

Pathogens commonly isolated from selected diseases

Bacteriology of Moderate-to-Severe Diabetic Foot Infections and In Vitro Activity of Antimicrobial Agents

Clostridium difficile Colitis

after trauma in children

BACTERIOLOGY PROFICIENCY TESTING PROGRAM

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

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

Mark Your Calendars Now! Next Event Ships: September 14, 2015

Cipro for gram positive cocci in urine

ESCHERICHIA COLI RESISTANCE AND GUT MICROBIOTA PROFILE IN PIGS RAISED WITH DIFFERENT ANTIMICROBIAL ADMINISTRATION IN FEED

Cleocin Pediatric clindamycin palmitate hydrochloride for oral solution, USP

CLEOCIN HCl Capsules contain clindamycin hydrochloride equivalent to 75 mg, 150 mg, or 300 mg of clindamycin.

Preventing Multi-Drug Resistant Organism (MDRO) Infections. For National Patient Safety Goal

2 0 hr. 2 hr. 4 hr. 8 hr. 10 hr. 12 hr.14 hr. 16 hr. 18 hr. 20 hr. 22 hr. 24 hr. (time)

Antibacterials. Recent data on linezolid and daptomycin

Multi-Drug Resistant Organisms (MDRO)

Ellie JC Goldstein Diane M. Citron C. Vreni Merriam Kerin L Tyrrell

In Vitro Activity of Netilmicin, Gentamicin, and Amikacin

Florida Health Care Association District 2 January 13, 2015 A.C. Burke, MA, CIC

Childrens Hospital Antibiogram for 2012 (Based on data from 2011)

In Vitro Susceptibility to Pexiganan of Bacteria Isolated from Infected Diabetic Foot Ulcers

6.0 ANTIBACTERIAL ACTIVITY OF CAROTENOID FROM HALOMONAS SPECIES AGAINST CHOSEN HUMAN BACTERIAL PATHOGENS

Running head: CLOSTRIDIUM DIFFICILE 1

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

available. and P. aeruginosa resistant to gentamicin by standardized disk testing (1) in the Microbiology Laboratory

6. STORAGE INSTRUCTIONS

03/09/2014. Infection Prevention and Control A Foundation Course. Talk outline

Transcription:

AAC Accepts, published online ahead of print on 19 December 2011 Antimicrob. Agents Chemother. doi:10.1128/aac.05655-11 Copyright 2011, 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 21 22 23 24 25 26 27 28 29 In vitro activity of CB-183,315, vancomycin and metronidazole against 556 strains of Clostridium difficile, 445 other intestinal anaerobes and 56 Enterobacteriaceae species 1 Diane M. Citron*, 1 Kerin L. Tyrrell, 1 C. Vreni Merriam and 1,2 Ellie J.C. Goldstein 1 R.M. Alden Research Lab, Culver City, CA 2 Geffen School of Medicine, UCLA, Los Angeles, CA Keywords: CB-183,315; Clostridium difficile; susceptibility; intestinal anaerobes Running title: CB-183,315 against C. difficile and intestinal organisms Corresponding author: Diane M. Citron R.M. Alden Research Lab 6133 Bristol Parkway Suite 175 Culver City, CA 90230 P 310-641-8340 F-310-641-8840 d.m.citron@att.net www.rmaldenresearch.com Abstract 1

30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 MICs for CB-183,315, a novel lipopeptide antibiotic, vancomycin and metronidazole were determined for intestinal anaerobes and Enterobacteriaceae. The MIC 90 for gram-negative anaerobes was >8192, 8192 and 4 µg/ml for CB-183,315, vancomycin and metronidazole, respectively. Against Enterobacteriaceae, MIC 90s were >8192 µg/ml, 1024 µg/ml and1024 µg/ml, respectively. CB-183,315 MIC 90 for C. difficile was 0.5 µg/ml. Its lack of activity against normal fecal organisms makes it a promising new agent for treating C. difficile. Clostridium difficile infection (CDI) is the most common cause of healthcare related diarrhea, usually associated with prior exposure to antimicrobial agents. The current theory is that the 2

53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 antibiotics disrupt the normal fecal microbiota, changing its complexity and diversity as a primary event, with the subsequent acquisition of toxigenic C. difficile as a secondary event in the development of the disease (6,11,14). CDI has increased in prevalence and severity during the last decade, resulting in increased mortality and complications, recurrent disease and prolonged hospital stay (7,17). Non-healthcare related cases of CDI are now being reported (7,16). Treatment options have been limited by unsatisfactory efficacy of current therapeutic agents, high recurrence rates of disease, disruption of normal intestinal microbiota, and colonization by VRE (1,2,15). Although a recently introduced macrocyclic antibiotic, fidaxomicin, shows equivalent primary cure rates with reduced recurrence rates compared to vancomycin (15) it shows equivalence against REA type BI strain associated disease. CB-183,315 is a novel lipopeptide antibiotic with gram-positive activity and is bactericidal against C. difficile. To study the spectrum of its activity, including effect on the normal components of fecal microbiota, we determined MICs of CB-183,315 and vancomycin against C. difficile and other intestinal anaerobes, plus E. coli, Klebsiella spp. and Enterobacter spp. For comparison, metronidazole was tested against the gram-negative anaerobes and the Enterobacteriaceae spp. The C. difficile strains were recent clinical isolates (2005 2008) recovered from stools of patients with CDI. Other isolates were from stool or infections containing organisms presumed to be of intestinal origin, identified by standard methods (13) and occasionally by 16S rdna sequence analysis (20), and stored in 20% skim milk at -70 o C. MIC values were determined by the agar dilution method according to CLSI procedures (4). Vancomycin and metronidazole laboratory standard powders were obtained from Sigma (St. 3

75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 Louis, MO) and CB-183,315 was provided by Cubist Pharmaceuticals, Inc. (Lexington, MA). All assay media for testing CB-183,315 were supplemented with a final concentration of 50 mg/l calcium, where Ca ++ concentrations were confirmed by Laboratory Specialists, Inc. (Westlake, Ohio). The Escherichia coli, Klebsiella pneumoniae and Enterobacter spp. strains were tested using Mueller Hinton agar, and incubated at 37 o C in ambient air, except for metronidazole, which was incubated in the anaerobic chamber (4). Quality control strains included Clostridium difficile ATCC 700057, Bacteroides fragilis ATCC 25285, Staphylococcus aureus ATCC 29213 and E. coli ATCC 25922 and were included each day of testing of the relevant set of test organisms. Time-kill studies were carried out on one restriction endonuclease analysis (REA) BI type (NAP1, ribotype 027) strain and one REA type Y (NAP4, ribotype 014) strain (3). CB-183,315 and vancomycin were prepared at 2, 4, and 8 times their MICs for each strain in supplemented Brucella broth. A drug-free growth control tube was included. Tubes were inoculated with ~ 10 6 CFU/ml, placed on a shaker and assayed at 0, 2, 4, 8, and 24h of incubation at 37 o C. Table 1 shows the range, MIC 50 and MIC 90 for the major groups of organisms. The CB-183,315 MIC range for C. difficile was 0.06 2.0 µg/ml with MIC 90 of 0.5 µg/ml; other gram-positive strains were inhibited by 0.03 16 µg/ml. The MIC 90 for CB-183,315 against the Bacteroides fragilis group, Prevotella spp., gram-negative cocci (Veillonella spp. and Acidaminococcus sp.), 4

98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 as well as E. coli, Enterobacter spp., and Klebsiella spp. was greater than 8192 µg/ml. Against fusobacteria, and Porphyromonas spp., the MIC 90 was 8192 µg/ml and 2048 µg/ml, respectively. In contrast, vancomycin had equally high MICs of >8192 µg/ml only against the fusobacteria and the gram-negative cocci; the B. fragilis group was inhibited by vancomycin at MIC 90 of 128 µg/ml (range 32-256 µg/ml), Porphyromonas by 4 µg/ml, and Prevotella spp. by 256 µg/ml of vancomycin. Vancomycin MICs ranged from 64 1024 µg/ml for E. coli and 256-1024 µg/ml for Enterobacter and Klebsiella species. Metronidazole was active against the gram-negative anaerobes at 0.06 to 8 µg/ml, but showed poor activity against the Enterobacteriaceae, with MICs ranging from 32 to1024 µg/ml. The time-kill studies showed that CB-183,315 at 4 and 8 times the MIC resulted in a 3 log 10 reduction in colony count after 24h incubation against both REA types tested. Vancomycin produced a similar result.. CB-183,315 reaches concentrations of 6494 + 3104 μg/g of feces after a 1g bid dose at day 5 of a 14 day course (Cubist Pharmaceuticals data on file) which is lower than the high MICs (>8192 µg/ml) demonstrated for Bacteroides and other groups of organisms of the intestinal anaerobes, thus CB-183,315 is likely to spare many of these important members of the normal microbiota. In contrast, vancomycin at a dose of 250 mg qid results in fecal levels that are generally above 2,000 μg/ml (12). Vancomycin thus has the potential for killing or inhibiting much of the normal aerobic and anaerobic fecal microbiota. Moreover, oral vancomycin was shown to decrease or suppress Bacteroides in volunteers while increasing occurrence of vancomycin-resistant enterococci (8,14). In contrast, fidaxomicin, a narrow spectrum 5

120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 macrocycle, displayed less impact on the Bacteroides populations in patients treated for CDI (14), which has been postulated as one reason for the associated lower relapse rate. Using temporal temperature gradient electrophoresis and fluorescent in situ hybridization and flow cytometry, Tannock et al.(19) compared the impact of fidaxomicin and vancomycin on seven phylogenetic groups, including clostridial clusters IV and XIVa, Bacteroides-Prevotella, Bifidobacterium, Atopobium, enterobacteria, and the Enterococcaceae-Lactobacillaceae group. Minimal changes were seen in these groups for the fidaxomicin treated patients while the vancomycin treated patients showed a marked decrease in Clostridium clusters IV and XIVa, Bacteroides and Bifidobacterium, and an increase in the Enterococcus-Lactobacillus and the enterobacteria groups. MICs for CB-183,315 against clostridia were similar to those reported for fidaxomicin (10), showing decreased activity against several species in Clostridium clusters XIVa, XVI, and XVIII, although unlike fidaxomicin, decreased activity of CB-183,315 was also present against several species in cluster I (5,9,18). MIC 90 for the C. clostridioforme group (cluster XIVa), was 16 µg/ml; for C. innocuum (cluster XVI) 4 µg/ml; for C. ramosum (Cluster XVIII) 8 µg/ml; and C. sphenoides, 1 strain, (cluster XIVa) 16 µg/ml. These findings suggest that CB-183,315 would show a similar lack of impact as fidaxomicin on the composition of the major groups of bowel microbiota. With its excellent activity against C. difficile and its narrower spectrum and lack of activity against other colonic aerobic and anaerobic microbiota, with the potential for fewer relapses, CB- 183,315 is a promising new drug treatment for CDI. 6

143 144 145 146 147 Acknowledgement This study was sponsored by a grant from Cubist Pharmaceuticals, Inc. We thank Eliza Leoncio for excellent technical assistance. Downloaded from http://aac.asm.org/ on October 1, 2018 by guest 7

148 Reference List 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 1. 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. 2. Bhalla, A., N. J. Pultz, A. J. Ray, C. K. Hoyen, E. C. Eckstein, and C. J. Donskey. 2003. Antianaerobic antibiotic therapy promotes overgrowth of antibiotic-resistant, gramnegative bacilli and vancomycin-resistant enterococci in the stool of colonized patients. Infect.Control Hosp.Epidemiol. 24:644-649. 3. Clabots, C. R., S. Johnson, K. M. Bettin, P. A. Mathie, M. E. Mulligan, D. R. Schaberg, L. R. Peterson, and D. N. Gerding. 1993. Development of a rapid and efficient restriction endonuclease analysis typing system for Clostridium difficile and correlation with other typing systems. J.Clin.Microbiol. 31:1870-1875. 4. Clinical and Laboratory Standards Institute. 2007. Methods for antimicrobial susceptibility testing of anaerobic bacteria, 7th ed. Approved standard M11-A7. Clinical and Laboratory Standards Institute, Wayne, PA. 5. Collins, M. D., P. A. Lawson, A. Willems, J. J. Cordoba, J. Fernandez-Garayzabal, P. Garcia, J. Cai, H. Hippe, and J. A. Farrow. 1994. The phylogeny of the genus 8

167 168 Clostridium: proposal of five new genera and eleven new species combinations. Int.J.Syst.Bacteriol. 44:812-826. 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 6. Dethlefsen, L., S. Huse, M. L. Sogin, and D. A. Relman. 2008. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rrna sequencing. PLoS.Biol. 6:e280. 7. Dubberke, E. R. and A. I. Wertheimer. 2009. Review of current literature on the economic burden of Clostridium difficile infection. Infect.Control Hosp.Epidemiol. 30:57-66. 8. Edlund, C., L. Barkholt, B. Olsson-Liljequist, and C. E. Nord. 1997. Effect of vancomycin on intestinal flora of patients who previously received antimicrobial therapy. Clin.Infect.Dis. 25:729-732. 9. Finegold, S. M., D. Molitoris, M. L. Vaisanen, Y. Song, C. Liu, and M. Bolanos. 2004. In vitro activities of OPT-80 and comparator drugs against intestinal bacteria. Antimicrob.Agents Chemother. 48:4898-4902. 10. Finegold, S. M., D. Molitoris, M. L. Vaisanen, Y. Song, C. Liu, and M. Bolanos. 2004. In vitro activities of OPT-80 and comparator drugs against intestinal bacteria. Antimicrob.Agents Chemother. 48:4898-4902. 9

184 185 11. Gerding, D. N. and S. Johnson. 2010. Management of Clostridium difficile infection: thinking inside and outside the box. Clin.Infect.Dis. 51:1306-1313. 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 12. 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. 13. Jousimies-Somer, H. R., P. Summanen, D. M. Citron, E. J. Baron, H. M. Wexler, and S. M. Finegold. 2002. Wadsworth-KTL Anaerobic Bacteriology Manual. Star Publishing, Belmont, CA. 14. Louie, T. J., J. Emery, W. Krulicki, B. Byrne, and M. Mah. 2009. OPT-80 eliminates Clostridium difficile and is sparing of bacteroides species during treatment of C. difficile infection. Antimicrob.Agents Chemother. 53:261-263. 15. Miller, M., L. Bernard, M. Thompson, D. Grima, and J. Pepin. 2010. Lack of increased colonization with vancomycin-resistant enterococci during preferential use of vancomycin for treatment during an outbreak of healthcare-associated Clostridium difficile infection. Infect.Control Hosp.Epidemiol. 31:710-715. 16. Rouphael, N. G., J. A. O'Donnell, J. Bhatnagar, F. Lewis, P. M. Polgreen, S. Beekmann, J. Guarner, G. E. Killgore, B. Coffman, J. Campbell, S. R. Zaki, and L. C. McDonald. 2008. Clostridium difficile-associated diarrhea: an emerging threat to pregnant women. Am.J.Obstet.Gynecol. 198:635-636. 10

203 204 17. Rupnik, M., M. H. Wilcox, and D. N. Gerding. 2009. Clostridium difficile infection: new developments in epidemiology and pathogenesis. Nat.Rev.Microbiol. 7:526-536. 205 206 207 208 209 210 211 212 213 214 215 216 18. Stackebrandt, E., I. Kramer, J. Swiderski, and H. Hippe. 1999. Phylogenetic basis for a taxonomic dissection of the genus Clostridium. FEMS Immunol.Med.Microbiol. 24:253-258. 19. Tannock, G. W., K. Munro, C. Taylor, B. Lawley, W. Young, B. Byrne, J. Emery, and T. Louie. 2010. A new macrocyclic antibiotic, fidaxomicin (OPT-80), causes less alteration to the bowel microbiota of Clostridium difficile-infected patients than does vancomycin. Microbiology 156:3354-3359. 20. Warren, Y. A., K. L. Tyrrell, D. M. Citron, and E. J. Goldstein. 2006. Clostridium aldenense sp. nov. and Clostridium citroniae sp. nov. isolated from human clinical infections. J.Clin.Microbiol. 44:2416-2422. 11

217 218 Table 1. In vitro activity of CB-183,315, Vancomycin and Metronidazole against Intestinal Organisms (μg/ml) CB-183,315 Vancomycin Metronidazole Group N Range MIC 50 MIC 90 Range MIC 50 MIC 90 Range MIC 50 MIC 90 Actinomyces species a 15 0.125-8.0 1.0 8.0 0.25 1.0 0.5 1.0 NT Bifidobacterium species b 14 0.06-2.0 0.5 2.0 0.25 2.0 0.5 1.0 NT Eggerthella lenta 17 1.0-16 4.0 8.0 1.0 2.0 2.0 2.0 NT Eubacterium limosum 13 0.06 1 0.25 0.5 1.0-2.0 2.0 2.0 NT Eubacterium group (other) c 35 0.06 8.0 0.25 2.0 0.25-32 1.0 2.0 NT Lactobacillus species, VAN-R d 20 0.5 2.0 1.0 2.0 >32 - >32 >32 >32 NT Lactobacillus species, VAN-S e 17 0.125 16.0 1.0 4.0 0.25 4.0 1.0 2.0 NT Propionibacterium species f 15 0.5-2.0 0.5 2.0 0.5 2.0 0.5 1.0 NT Clostridium clostridioforme NT grp g (CL. XIVa) 20 1.0-16.0 8.0 16.0 0.5 1.0 1.0 1.0 Clostridium difficile (Cl. XI) 556 0.06-2 0.5 0.5 0.25 4.0 1.0 2.0 NT Clostridium innocuum (Cl.XV) 22 1.0 4.0 2.0 4.0 8.0 16.0 8.0 16.0 NT Clostridium perfringens (Cl.I) 20 0.25-1.0 0.5 1.0 0.5-1 0.5 1 NT Clostridium ramosum (Cl. NT XVIII) 20 2.0-8.0 4.0 8.0 2-8 4 8 Clostridium species (other) h 39 0.125-16.0 1.0 16.0 0.25-32 1 2 NT Anaerobic gram-positive cocci i 49 <0.03-4.0 0.25 0.5 0.06-1 0.25 0.5 NT Bacteroides fragilis group j 21 8192->8192 8192 >8192 32-256 64 128 0.25-2 1 2 128- Fusobacterium species k 20 128->8192 512 8192 >8192 512 >8192 0.06-1.25.5 Porphyromonas species l 21 64-8192 1024 2048 1-32 4 4 0.06-1 0.25 1 Prevotella species m 20 8192->8192 8192 >8192 32-512 128 256 0.25-4 1 4 256- Gram-negative cocci n 21 8192->8192 >8192 >8192 >8192 2048 >8192 0.06-8 2 4 >8192 >8192 128- E. coli 18 4096->8192 64-256 128 256 1024 256 512 12

219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 >8192- >8192 >8192 Enterobacter species o 18 >8192 256-1024 512 1024 32-1024 512 1024 >8192 >8192 >8192 128- Klebsiella species p 20 256-1024 1024 1024 1024 512 1024 Cl, phylogenetic cluster a Actinomyces israelii (2), A. meyeri (2), A. naeslundii (1), A. neuii ss anitratus (2), A. neuii ss neuii (1), A. odontolyticus (2), A. turicensis (2), and A. viscosus (3). b Bifidobacterium adolescentis (2), B. bifidum (1), B. breve (4), B. catenulatum (2), B. dentium (1), B. longum (2), B. pseudocatenulatum (1), and B. urinalis (1). c Collinsella aerofaciens (6), Eubacterium biforme (1), E. contortum (1), E. cylindroides (1), E. timidum (3), Pseudoramibacter alactolyticum (4) and Eubacterium species (19). d Lactobacillus brevis (1), L. casei (7), L. fermentum (2), L. rhamnosus (6), and Lactobacillus GG (4). e Atopobium parvulum (1), Lactobacillus acidophilus (2), L. catenaformis (3), L. crispatus (1), L. gasseri (3), L. iners (1), L. jensenii (4), and L. leichmannii (2 ) f Propionibacterium acnes (7), P. avidum (4), and P. granulosum (4). g Clostridium aldenense (4), C. bolteae (5), C. citroniae (3), C. clostridioforme (2), and C. hathewayi (6). h Clostridium bifermentans (1), C. butyricum (5), C. cadaveris (3), C. cochlearium (2), C. fallax (1), C. glycolicum (3), C. hastiforme (1), C. hylemonae (1), C. indolis (1), C. leptum (2), C. nexile (1), C. paraputrificum (4), C. sartagoforme (1), C. scindens (1), C. sordelli (2), C. sphenoides (1), C. sporogenes (2), C. subterminale (1), C. symbiosum (4), and C. tertium (2). 13

240 241 242 243 244 245 246 247 248 249 250 251 252 253 i Anaerococcus lactolyticus (2), A. octavius (1), A. prevotii (6), A. tetradius (5), A. vaginalis (4) ; Finegoldia magna (7) ; Peptoniphilus asaccharolyticus (5), P. harei (2), P. indolicus (2), P. ivorii (3), and P. lacrimalis (2); Peptostreptococcus anaerobius (5) and Parvimonas micra (5). j Bacteroides caccae (1), B. distasonis (1), B. fragilis (11), B. nordii (1), B. ovatus (3), B. thetaiotaomicron (3), B. vulgatus (1). k Fusobacterium gonidiaformans (1), F. mortiferum-varium group (6), F. necrophorum (2), F. nucleatum (10), and F. russii (1). l Porphyromonas asaccharolytica (9), P. gingivalis (4), P. somerae (6), P. uenonis (2). m Prevotella bivia (10), P. disiens (3), P. intermedia/nigrescens (1), P. loeschii (3), and P. melaninogenica (3). n Acidaminococcus fermentans (8), Veillonella species (13). o Enterobacter aerogenes (6), Enterobacter cloacae (11), Enterobacter sp. (1) p Klebsiella oxytoca (3), Klebsiella pneumoniae (17) 14