.'URRENT THERAPEUTIC RESEA. VOLUME 66, NUMBER 3, MAY/JuNE 2005

Similar documents
Reiner Schaumann, 1 Ellie J. C. Goldstein, 2 Jochen Forberg 3 and Arne C. Rodloff 1

Tel: Fax:

SESSION XVI NEW ANTIBIOTICS

Scottish Medicines Consortium

Effects of Minocycline and Other Antibiotics on Fusobacterium necrophorum Infections in Mice

Therapeutic Efficacy of 29 Antimicrobial Regimens in Experimental Intraabdominal Sepsis

Risk of Infection Following Penetrating Abdominal Trauma: A Selective Review

VOL. XXIII NO. II THE JOURNAL OF ANTIBIOTICS 559. ANTIBIOTIC 6640.* Ill

on February 12, 2018 by guest

Antimicrobial Pharmacodynamics

Appropriate antimicrobial therapy in HAP: What does this mean?

Antibiotic Updates: Part II

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

a. 379 laboratories provided quantitative results, e.g (DD method) to 35.4% (MIC method) of all participants; see Table 2.

Moxifloxacin resistance is prevalent among Bacteroides and Prevotella species in Greece

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

Multicenter Study of In Vitro Susceptibility of the Bacteroides fragilis Group, 1995 to 1996, with Comparison of Resistance Trends from 1990 to 1996

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

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

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

Clinical Policy: Clindamycin (Cleocin) Reference Number: CP.HNMC.08 Effective Date: Last Review Date: Line of Business: Medicaid - HNMC

Secondary peritonitis

Pharmacological Evaluation of Amikacin in Neonates

Introduction to Pharmacokinetics and Pharmacodynamics

Animal models and PK/PD. Examples with selected antibiotics

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)

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

Comparison of Clindamycin, Erythromycin, and Methicillin in Streptococcal Infections in Monkeys

Streptococcus pneumoniae Response to Repeated Moxifloxacin or Levofloxacin Exposure in a Rabbit Tissue Cage Model

Give the Right Antibiotics in Trauma Mitchell J Daley, PharmD, BCPS

Pinni Meedha Mojutho Ammanu Dengina Koduku Part 1 Kama Kathalu

OPTIMIZATION OF PK/PD OF ANTIBIOTICS FOR RESISTANT GRAM-NEGATIVE ORGANISMS

Antibiotics & treatment of Acute Bcterial Sinusitis. Walid Reda Product Manager. Do your antimicrobial options meet your needs?

General Infectious Disease Concepts/Resources

Comparison of BD BACTEC Plus Blood Culture Media versus VersaTREK REDOX

The pharmacological and microbiological basis of PK/PD : why did we need to invent PK/PD in the first place? Paul M. Tulkens

against Clinical Isolates of Gram-Positive Bacteria

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

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

See Important Reminder at the end of this policy for important regulatory and legal information.

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

Antibiotic Abyss. Discussion Points. MRSA Treatment Guidelines

Journal of Antimicrobial Chemotherapy Advance Access published August 26, 2006

PIPERACILLIN- TAZOBACTAM INJECTION - SUPPLY PROBLEMS

Background and Plan of Analysis

Reduce the risk of recurrence Clear bacterial infections fast and thoroughly

DETERMINING CORRECT DOSING REGIMENS OF ANTIBIOTICS BASED ON THE THEIR BACTERICIDAL ACTIVITY*

Alasdair P. MacGowan,* Chris A. Rogers, H. Alan Holt, and Karen E. Bowker

JAC Bactericidal index: a new way to assess quinolone bactericidal activity in vitro

PDF hosted at the Radboud Repository of the Radboud University Nijmegen

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

CLINICAL USE OF AMINOGLYCOSIDES AND FLUOROQUINOLONES

ESCMID Online Lecture Library. by author

Measure Information Form

USA Product Label LINCOCIN. brand of lincomycin hydrochloride tablets. brand of lincomycin hydrochloride injection, USP. For Use in Animals Only

These recommendations were approved for use by the Pharmaceutical and Therapeutics Committee, RCWMCH on 1 February 2017.

Clinical Usefulness of Multi-facility Microbiology Laboratory Database Analysis by WHONET

Antibiotic Usage Guidelines in Hospital

Who should read this document? 2. Key practice points 2. Background/ Scope/ Definitions 2. What is new in this version? 3

Initial Management of Infections in the Era of Enhanced Antimicrobial Resistance

Educating Clinical and Public Health Laboratories About Antimicrobial Resistance Challenges

Combination antibiotic therapy: comparison of constant infusion and intermittent bolus dosing in an experimental animal model

Original Article. Ratri Hortiwakul, M.Sc.*, Pantip Chayakul, M.D.*, Natnicha Ingviya, B.Sc.**

Alasdair P. MacGowan*, Mandy Wootton and H. Alan Holt

Antibiotic. Antibiotic Classes, Spectrum of Activity & Antibiotic Reporting

ESCMID Online Lecture Library. by author

Antibiotic Updates: Part I

What does multiresistance actually mean? Yohei Doi, MD, PhD University of Pittsburgh

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

coverage. The purpose of this study was to compare the susceptibility of 297 strains of anaerobic and a new broad-spectrum semisynthetic

Synergism Between Penicillin, Clindamycin, or Metronidazole and Gentamicin Against Species of the Bacteroides melaninogenicus and

THERAPY OF ANAEROBIC INFECTIONS LUNG ABSCESS BRAIN ABSCESS

April 25, 2018 Edited by: Gregory K. Perry, PharmD, BCPS-AQID

The Basics: Using CLSI Antimicrobial Susceptibility Testing Standards

Antibacterial Resistance: Research Efforts. Henry F. Chambers, MD Professor of Medicine University of California San Francisco

Scottish Medicines Consortium

Role of moxifloxacin for the treatment of commmunity-acquired complicated intra-abdominal infections in Taiwan

Int.J.Curr.Microbiol.App.Sci (2017) 6(3):

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

Effeet on Bacterial Growth

CHSPSC, LLC Antimicrobial Stewardship Education Series

Discover the. Discover the. innovative science. Veraflox (pradofloxacin) Veraflox. Efficacy. Safety. Ease-of-use.

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

Persistent in Kidneys

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

Case 2 Synergy satellite event: Good morning pharmacists! Case studies on antimicrobial resistance

Antimicrobial Prophylaxis in the Surgical Patient. M. J. Osgood

Summary of unmet need guidance and statistical challenges

Intrinsic, implied and default resistance

ESCMID Online Lecture Library. by author

Diabetic Foot Infection. Dr David Orr Consultant Microbiologist Lancashire Teaching Hospitals

Period of study: 12 Nov 2002 to 08 Apr 2004 (first subject s first visit to last subject s last visit)

Successful stewardship in hospital settings

Implantation of Tissue Chambers in Turkeys: A Pilot Study

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

Preserving bacterial susceptibility Implementing Antimicrobial Stewardship Programs Debra A. Goff, Pharm.D., FCCP

11/10/2016. Skin and Soft Tissue Infections. Disclosures. Educational Need/Practice Gap. Objectives. Case #1

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.

Principles of Infectious Disease. Dr. Ezra Levy CSUHS PA Program

Proceedings of the 13th International Congress of the World Equine Veterinary Association WEVA

Transcription:

.'URRENT THERAPEUTIC RESEA VOLUME 66, NUMBER 3, MAY/JuNE 2005 Efficacy of Moxifloxacin Monotherapy Versus Gatifloxacin Monotherapy, Piperacillin- Tazobactam Combination Therapy, and Clindamycin Plus Gentamicin Combination Therapy: An Experimental Study in a Rat Model of Intra-Abdominal Sepsis Induced by Fluoroquinolone-Resistant Bacteroides fragilis Ronald L. Cisneros, MS; and Andrew B. Onderdonk, PhD Channing Laboratory, Brigham and Women's Hospital, Boston, Massachusetts; and Department of Pathology, Harvard Medical School, Boston, Massachusetts ABSTRACT Background: In intra-abdominal infections, the activity of antimicrobial agents against Bacteroides fragilis and phenotypically related organisms, and the increasing resistance of these organisms, are of particular importance and concern to surgeons: In vitro data suggest that moxifloxacin is more active than other quinolones against obligately anaerobic organisms, including Bacteroides spp. Objective: The aim of this study was to compare the efficacy of moxifloxacin monotherapy versus gatifloxacin monotherapy and 2 combination therapies (piperacillin-tazobactam and clindamycin plus gentamicin) in a rat model of intra-abdominal sepsis. The end point was marked by the incidence of mortality and intra-abdominal abscesses at necropsy 7 days after bacterial challenge. Methods: Three different strains of B fragilis with different degrees of resistance to moxifloxacin (minimum inhibitory concentrations [MICs]: 4, 8, and 16 pg/ml) were added to the challenge inoculum in 3 separate experiments. Groups of 20 animals were used in each experiment. Group 1 served as salinetreated controls; group 2 received moxifloxacin 15 mg QD; group 3 received gatifloxacin 25 mg QD; group 4 received piperacillin-tazobactam 93 mg (~83 mg of piperacillin) QD; and group 5 received a combination of clindamycin 15 mg TID plus gentamicin 2 mg TID. All treatments were given intramuscularly. For all antimicrobials, dose was based on peak and trough serum drug concentrations determined by prior testing, with animal doses adjusted based on the ratio of body surface area to body weight, and comparing these doses and levels with studies in humans. Accepted for publication April 26, 2005. Reproduction in whole or part is not permitted. doi:l 0.1016/j.curtheres.2005.06.003 0011-393X/05/$19.00 222 Copyright 2005 Excerpta Medica, Inc.

R.L. Cisneros and A.B. Onderdonk Results: In all 3 experiments, the mortality rate with moxifloxacin was significantly lower or statistically similar compared with antibiotic active comparatots (P _< 0.024). In addition, there were no significant differences in the incidence of abscess with moxifloxacin versus its comparators or between the 3 moxifloxacin groups across experiments. The best results for moxifloxacin were found in the experiment in which the B fragilis strain with MIC 16 pg/ml was added to the inoculum. Conclusion: The results of this study in an animal model of intra-abdominal sepsis induced by fluoroquinolone-resistant B fragilis suggest that moxifloxacin monotherapy performs as well as combination regimens such as piperacillintazobactam and clindamycin plus gentamicin, and is as effective as other fluoroquinolones with antianaerobic activity, such as gatifloxacin. (Curr Ther Res Clin Exp. 2005;66:222-229) Copyright 2005 Excerpta Medica, Inc. Key words: intra-abdominal sepsis, resistance, Bacteroides, quinolone. INTRODUCTION Previous studies from the Channing Laboratory, Brigham and Women's Hospital, Boston, Massachusetts, 1,2 have shown a variety of antimicrobial agents to be effective in preventing both mortality and abscess formation in animal models of intra-abdominal sepsis. 3 The experimental disease process consists of 2 phases-- early acute peritonitis and the development of abscesses in recipients surviving a fecal inoculum. The 2 phases of intra-abdominal sepsis are associated with distinctly different bacterial populations. During early acute peritonitis, Escherichia coli and other aerobic gram-negative organisms are numerically dominant and appear to be the cause of mortality. The second, more chronic stage of the disease, abscess formation, requires the presence of obligate anaerobes, such as Bacteroides fragilis. 3 In the rat model of intra-abdominal sepsis, use of antimicrobial agents active against the coliform population has been shown to reduce acute mortality, while antimicrobial agents active against obligate anaerobes have been shown to reduce the incidence of subsequent abscess formation. 1,2 In intra-abdominal infections, the activity of an antimicrobial agent against B fragilis and phenotypically related members of this group is of particular importance. In vitro assays suggest that agents lacking documented activity against this organism often do not perform well in experimental models simulating intra-abdominal sepsis or in humans with serious abdominal infections. 4,5 Other in vitro susceptibility testing studies have suggested that some strains of B fragilis are moderately resistant to moxifloxacin. 6,7 However, it is not known whether this resistance was an artifact of the testing methodology, or whether these observations were clinically relevant. To determine the in vivo relevance of these in vitro observations, we used a rat model of intra-abdominal sepsis to evaluate 3 strains of B fragilis for which the minimum inhibitory concentrations (MICs) for moxifloxacin were 4, 8, and 16 pg/ml. For this group of organisms, the minimum concentration of moxifloxacin that was inhibitory for 90% of all isolates was 0.25 to 2.00 pg/ml. 223

CURRENT THERAPEUTIC RESEARCH MATERIALS AND METHODS Animals Male virus antibody-free (VAF) Wistar rats weighing 175 to 200 g were used for the study (Charles River Laboratories, Wilmington, Massachusetts). All animals were housed in a VAF animal facility and maintained under strict Association for Assessment and Accreditation of Laboratory Animal Care International and National Institutes of Health guidelines for the care and use of laboratory animals. 8,9 Inoculum Bacteriology The mixed aerobe-anaerobe inoculum from meat-fed rats was prepared as described previously. 3 Quantitative and qualitative bacteriology of this inoculum was performed to ensure the presence of appropriate microbial species. All procedures were carried out in an anaerobic chamber to ensure that organisms in the inoculum would remain viable and representative of the original cecal material. Enterobacteriaceae were identified using the Vitek system (biom~rieux SA, Durham, North Carolina); anaerobic isolates were identified using the Microbial Identification System (Microbial ID, Inc., Newark, Delaware); and aerobic isolates were identified using established methods. 1 All isolate counts were recorded as log10 colony-forming units per milliliter (CFU/mL) of inoculum. Resistant Bacteroides fragilis Three clinical strains of B fragilis (Bayer Corporation, West Haven, Connecticut) with different levels of resistance to moxifloxacin were used in the experiments. Each strain was grown in brain-heart infusion-supplemented broth for 24 to 48 hours, aliquoted, flash-frozen in liquid nitrogen, and stored at -80 C. The MIC of moxifloxacin for each strain was confirmed using a microbroth dilution. The MICs for strains 200302049, 200211034, and 200205127 were 4, 8, and 16 pg/ml, respectively. Before each experiment, the aliquots of the test strain were thawed, and quantitative counts were performed to document the concentration of viable organisms. Viability is maintained at a constant level for up to 2 years after flash freezing. The test strains were then added individually to the inoculum, for a final concentration of 1 108 CFU/mL per animal. Implantation of the Inoculum Animals were anesthetized with pentobarbital 60 mg/kg and prepared for surgical implantation of the inoculum. Their abdomens were shaved, and an iodine solution was applied. A 1-cm midline incision was made through the skin and anterior abdominal wall, and a gelatin capsule containing 0.5 ml of the inoculum containing 10% wt/vol barium sulfate was inserted into the pelvic region, as described previously. 3 Although the barium sulfate is nonabscessogenic alone, it acts as a nidus for abscess formation. The incision was closed with 3-0 silk suture, and the animals were returned to their cages and monitored every 8 hours for the duration of the 7-day experiment. 224

R.L. Cisneros and A.B. Onderdonk Experimental Design The design of each of the 3 experiments was identical. Five groups of 20 animals were evaluated in this study. Group 1 served as saline-treated controls; group 2 received moxifloxacin 15 mg IM QD; group 3 received gatifloxacin 25 mg IM QD; group 4 received a combination solution of piperacillin plus tazobactam 93 mg IM QD; and group 5 received combination therapy with clindamycin 15 mg TID plus gentamicin 2 mg TID IM. For all antimicrobials, dose was based on peak and trough serum drug concentrations determined by prior testing, with animal doses adjusted based on the ratio of body surface area to body weight, and comparing these doses and levels with studies in humans. The mean Cma x values were 6.0, 2.5, 86, and 5.5/6.9 pg/ml in animals receiving moxifloxacin (n = 5), gatifloxacin, piperacillin-tazobactam, and clindamycin plus gentamicin, respectively. All Cma ~ values were consistent with those obtained in therapeutic use of these agents in humans. 11,12 Treatment was initiated within 2 hours of surgical implantation of the cecal inoculum and was continued for 7 days at intervals appropriate for the various antimicrobial agents. Cma ~ values were determined using hemolysis inhibition assay 13 1 hour after study drug administration. Evaluation of Animals Animals were monitored every 8 hours for mortality. Moribund animals were euthanized using carbon dioxide (CO2) and were included in the mortality analysis but not examined for abscess development. Seven days after bacterial challenge and initiation of antibiotic treatment, surviving animals were euthanized using CO 2, and their abdominal cavities were examined for the presence of intraabdominal abscesses. These examinations were blinded, and abscesses were considered to be present if loculated collections of purulent material containing polymorphonuclear cells and bacteria were detected) Culture and microscopic examination of exudates visually confirmed the presence of abscesses. Statistical Analysis Mortality and the incidence of abscesses in surviving animals were compared between the control and treated groups and between the treated groups. The program used a 2 x 2 contingency table using the Fisher exact test to provide a 1-sided P value (Instat version 3.0, GraphPad Software, Inc., San Diego, California). P < 0.05 was considered statistically significant. This study was powered to detect a 35% difference in mortality and a 50% difference in abscess incidence versus controls, with 95% confidence. RESULTS For B fragilis 200302049 (Tables I and li), 200211034 (Tables III and IV), and 200205127 (Tables V and VI), all treatment groups had significantly higher mortality rates compared with untreated controls (all, P < 0.05). In all 3 experiments, the mortality rate with moxifloxacin was statistically similar compared 225

CURRENT THERAPEUTIC RESEARCH Table I. Mortality rate and incidence of abscess in rats challenged with an inoculum enhanced with Bacteroides fragilis 200302049.* Mortality Rate t Incidence of Abscess P vs P vs P vs Group No. (%) Control No. (%) Control Moxifloxacin Saline (control) 10/19 (53) - 9/9 (100) - - Moxifloxacin 3/19 (16) <0.020 4/16 (25) <0.001 - Gatifloxacin 2/19 (11) <0.007 2/17 (12) <0.001 <0.394 Piperacillin/tazobactarn 3/19 (16) <0.020 2/16 (13) <0.001 <0.654 Clindamycin + gentamicin 2/19 (11) <0.007 1/17 (6) <0.001 <0.175 *Minimum inhibitory concentration, 4 IJg/mL moxifloxacin. tno significant differences versus moxifloxacin were found. Table II. Isolates recovered from abdominal abscesses in rats challenged with an inoculum enhanced with Bacteroides fragilis 200302049.* Run 1 Run 2 Run 3 Group Isolate MIC Isolate MIC Isolate MIC Saline (control) B fragilis <1 B fragilis <1 B fragilis 32 Moxifloxacin B fragilis 16 B fragilis 16 B fragilis - Gatifloxacin B fragilis 16 B fragilis 16 B fragilis - Piperacillin/tazobactam B fragilis 8 B fragilis 8 B fragilis 8 *Minimum inhibitory concentration (MiC), 4 IJg/mL moxifloxacin. Table III. Mortality rate and incidence of abscess in rats challenged with an inoculum enhanced with Bacteroides fragilis 200211034.* Mortality Rate t Incidence of Abscess P vs P vs P vs Group No. (%) Control No. (%) Control Moxifloxacin Saline (control) 11/20 (55) - 9/9 (100) - - Moxifloxacin 4/20 (20) <0.025 4/16 (25) <0.187 - Gatifloxacin 2/20 (10) <0.003 3/18 (17) <0.063 <0.682 Piperacillin/tazobactam 3/19 (16) <0.013 3/16 (19) <0.096 1.000 Clindamycin + gentamicin 2/20 (10) <0.003 2/18 (11) <0.025 <0.388 *Minimum inhibitory concentration, 8 pg/ml moxifloxacin. tno significant differences versus moxifloxacin were found. 226

R.L. Cisneros and A.B. Onderdonk Table IV. Isolates recovered from abdominal abscesses in rats challenged with an inoculum enhanced with Bacteroides fragilis 200211034.* Run 1 Run 2 Group Isolate MIC Isolate MIC Saline (control) B fragilis 4 B fragilis 2 Moxifloxacin B fragilis 8 B fragilis 16 Gatifloxacin Bacteroides oralis 32 - - Piperacillin/tazobactam Bacteroides spp 8 B fragilis 16 Clindamycin + gentamicin B fragilis 8 - - *Minimum inhibitory concentration (MIC), 8 pg/ml moxifloxacin. Table V. Mortality rate and incidence of abscess in rats challenged with an inoculum enhanced with Bacteroides fragilis 200205127.* Mortality Rate t Incidence of Abscess P vs P vs P vs Group No. (%) Control No. (%) Control Moxifloxacin Saline (control) 10/19 (53) - 9/9 (100) - - Moxifloxacin 0/20 (0) <0.001 2/19 (11) <0.001 - Gatifloxacin 0/20 (0) <0.001 5/20 (25) <0.001 <0.408 Piperacillin/tazobactam 0/19 (0) <0.001 2/19 (11 ) <0.001 <1.397 Clindamycin + gentamicin 0/20 (0) <0.001 2/18 (11) <0.001 1.000 *Minimum inhibitory concentration, 16 Mg/mL moxifloxacin. tno significant differences versus moxifloxacin were found. Table VI. Isolates recovered from abdominal abscesses in rats challenged with an inoculum enhanced with Bacteroidesfragilis 200205127.* Run 1 Run 2 Run 3 Group Isolate MIC Isolate MIC Isolate MIC Saline (control) B fragilis 8 B fragilis 8 - - Moxifloxacin Bacteroides spp 2 B fragilis 16 B fragilis 16 Gatifloxacin Bacteroides spp 16 B fragilis 8 - - Piperacillin/tazobactam B fragilis 8 B fragilis 8 - - Clindamycin + gentamicin B fragilis 16 B fragilis 8 - - *Minimum inhibitory concentration (MIC), 16 IJg/mL moxifloxacin. 227

CURRENT THERAPEUTIC RESEARCH with antibiotic active comparators. When the incidence of abscess at necropsy was compared between all of the treatment groups, the rates were statistically similar (Tables I, III, and V). Analysis of abscess cultures indicated that the organisms recovered were consistent with inoculum contents and did not conflict with the results of the analysis of organisms and their susceptibility data (Tables II, IV, and VI). DISCUSSION Previous in vitro susceptibility testing studies have suggested that some strains of B fragilis are moderately resistant to moxifioxacin, 6 but it was not known whether these findings were an artifact of the testing methodology or whether these observations were clinically relevant. To determine the relevance of these in vitro observations in vivo, we used a rat model of intra-abdominal sepsis to evaluate 3 strains of B fragilis for which the MICs for moxifloxacin ranged from 4 to 16 pg/ml. In all 3 experiments, the mortality rate in the moxifloxacin group was statistically similar to or lower than that in the groups receiving comparator antibiotics (gatifloxacin, piperacillin/tazobactam, and clindamycin plus gentamicin). In addition, there were no statistically significant differences in abscess rates with moxifloxacin either compared with the other antibiotics tested or between experiments for moxifloxacin. Indeed, the best results for moxifloxacin were found in the experiment in which the inoculum contained the B fragilis strain for which the MIC was 16 pg/ml. CONCLUSION In this study in a rat model of intra-abdominal sepsis, moxifloxacin used as monotherapy performed as well as the combination regimens piperacillin-tazobactam and clindamycin plus gentamicin, and was as effective as gatifloxacin, another fluoroquinolone with antianaerobic activity. ACKNOWLEDGMENT This study was financially supported by an unrestricted grant from Bayer Corporation. REFERENCES 1. Cisneros RL, Bawdon RE, Onderdonk AB. Efficacy of ampicillin/sulbactam for the treatment of experimental intra-abdominal sepsis. Curr TherRes Clin Exp. 1990;48:1021-1029. 2. Bartlett JG, Louis T J, Gorbach SL, Onderdonk AB. Therapeutic efficacy of 29 antimicrobial regimens in experimental intraabdominal sepsis. Rev InfectDis. 1981;3:535-542. 3. Weinstein WM, Onderdonk AB, Bartlett JG, Gorbach SL. Experimental intra-abdominal abscesses in rats: Development of an experimental model. Infect Immun. 1974;10: 1250-1255. 228

R.L. Cisneros and A.B. Onderdonk 4. Thadepalli H, Chuah SK, Gollapudi S. Therapeutic efficacy of moxifloxacin, a new quinolone, in the treatment of experimental intra-abdominal abscesses induced by Bacteroides fragilis in mice. Chemotherapy. 2004;50:76-80. 5. Xuan D, Zhong M, Mattoes H, et al. Streptococcus pneumoniae response to repeated moxifloxacin or levofloxacin exposure in a rabbit tissue cage model. Antimicrob Agents Chemother. 2001;45:794-799. 6. Snydman DR, Jacobus NV, McDermott LA, et al. In vitro activities of newer quinolones against bacteroides group organisms [published correction appears in Antimicrob Agents Chemother. 2003;47:831]. Antimicrob Agents Chemother. 2002;46:3276-3279. 7. Hedberg M, Nord CE, for the ESCMID Study Group on Antimicrobial Resistance in Anaerobic Bacteria. Antimicrobial susceptibility of Bacteroides fragilis group isolates in Europe. Clin Microbiol Infect. 2003;9:475-488. 8. Bayne K, for the American Physiological Society. Revised guide for the care and use of laboratory animals. Physiologist. 1996;39:199, 208-211. 9. Institutional Animal Care and Use Committee Guidebook. 2nd ed. Bethesda, Md: National Institutes of Health; 2002. 10. Murray PR. Manual of Clinical Microbiology. 8th ed. Washington, DC: American Society for Microbiology Press; 2004. 11. Goldstein EJ. Intra-abdominal anaerobic infections: Bacteriology and therapeutic potential of newer antimicrobial carbapenem, fluoroquinolone, and desfluoroquinolone therapeutic agents. Clin Infect Dis. 2002;35(Suppl 1):$106-$111. 12. Zhanel GG, Ennis K, Vercaigne L, et al. A critical review of the fluoroquinolones: Focus on respiratory infections [published correction appears in Drugs. 2002;62: 944]. Drugs. 2002;62:13-59. 13. Louie T J, Tally FP, Bartlett JG, Gorbach SL. Rapid microbiological assay for chloramphenicol and tetracyclines. Antimicrob Agents Chemother. 1976;9:874-878. Address correspondence to: Ronald L. Cisneros, MS, Channing Laboratory, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115. E-mail: rcisneros@rics.bwh.harvard.edu 229