In vitro pharmacodynamics of colistin against Acinetobacter baumannii clinical isolates

Similar documents
Received 10 November 2006/Returned for modification 9 January 2007/Accepted 17 July 2007

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

Tel: Fax:

ETX2514SUL (sulbactam/etx2514) for the treatment of Acinetobacter baumannii infections

Burton's Microbiology for the Health Sciences. Chapter 9. Controlling Microbial Growth in Vivo Using Antimicrobial Agents

Doripenem: A new carbapenem antibiotic a review of comparative antimicrobial and bactericidal activities

Summary of the latest data on antibiotic resistance in the European Union

Appropriate antimicrobial therapy in HAP: What does this mean?

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

Antimicrobial Cycling. Donald E Low University of Toronto

Antimicrobial Pharmacodynamics

Influence of ph on Adaptive Resistance of Pseudomonas aeruginosa to Aminoglycosides and Their Postantibiotic Effects

Effect of pulmonary surfactant on antimicrobial activity in-vitro

Journal of Antimicrobial Chemotherapy Advance Access published August 26, 2006

Activity of a novel aminoglycoside, ACHN-490, against clinical isolates of Escherichia coli and Klebsiella pneumoniae from New York City

Other Beta - lactam Antibiotics

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

Outline. Antimicrobial resistance. Antimicrobial resistance in gram negative bacilli. % susceptibility 7/11/2010

DRUG-RESISTANT ACINETOBACTER BAUMANNII A GROWING SUPERBUG POPULATION. Cara Wilder Ph.D. Technical Writer March 13 th 2014

Multi-drug resistant Acinetobacter (MDRA) Surveillance and Control. Alison Holmes

Dynamic Drug Combination Response on Pathogenic Mutations of Staphylococcus aureus

Update on Resistance and Epidemiology of Nosocomial Respiratory Pathogens in Asia. Po-Ren Hsueh. National Taiwan University Hospital

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

Multidrug-Resistant Organisms: How Do We Define them? How do We Stop Them?

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

Other β-lactamase Inhibitor (BLI) Combinations: Focus on VNRX-5133, WCK 5222 and ETX2514SUL

Isolation of Urinary Tract Pathogens and Study of their Drug Susceptibility Patterns

R-factor mediated trimethoprim resistance: result of two three-month clinical surveys

ETX2514: Responding to the global threat of nosocomial multidrug and extremely drug resistant Gram-negative pathogens

MICRONAUT MICRONAUT-S Detection of Resistance Mechanisms. Innovation with Integrity BMD MIC

COMMITTEE FOR VETERINARY MEDICINAL PRODUCTS

PDF hosted at the Radboud Repository of the Radboud University Nijmegen

New Drugs for Bad Bugs- Statewide Antibiogram

CF WELL Pharmacology: Microbiology & Antibiotics

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

Advance Access published September 16, 2004

These life-saving drugs have been a boon to medical care and benefited hundreds of million patients around the globe.

The Disinfecting Effect of Electrolyzed Water Produced by GEN-X-3. Laboratory of Diagnostic Medicine, College of Medicine, Soonchunhyang University

Global Antibiotic Research and Development (GARD) Partnership Pasteur Institute DNDi Meeting February 29, 2016

Pharmacological Evaluation of Amikacin in Neonates

Available online at ISSN No:

The First Report of CMY, AAC(6')-Ib and 16S rrna Methylase Genes among Pseudomonas aeruginosa Isolates from Iran

ETX0282, a Novel Oral Agent Against Multidrug-Resistant Enterobacteriaceae

The Basics: Using CLSI Antimicrobial Susceptibility Testing Standards

Detection and Quantitation of the Etiologic Agents of Ventilator Associated Pneumonia in Endotracheal Tube Aspirates From Patients in Iran

Prevalence of Metallo-Beta-Lactamase Producing Pseudomonas aeruginosa and its antibiogram in a tertiary care centre

DR. MICHAEL A. BORG DIRECTOR OF INFECTION PREVENTION & CONTROL MATER DEI HOSPITAL - MALTA

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

UDC: : :579.22/ :615.28

Postantibiotic effect of aminoglycosides on Gram-negative bacteria evaluated by a new method

Microbiology ( Bacteriology) sheet # 7

Testimony of the Natural Resources Defense Council on Senate Bill 785

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

Mechanism of antibiotic resistance

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

Principles of Antimicrobial therapy

Overnight identification of imipenem-resistant Acinetobacter baumannii carriage in hospitalized patients

MDR Acinetobacter baumannii. Has the post antibiotic era arrived? Dr. Michael A. Borg Infection Control Dept Mater Dei Hospital Malta

International Journal of Advances in Pharmacy and Biotechnology Vol.3, Issue-2, 2017, 1-7 Research Article Open Access.

Pornpan Koomanachai a, Surapee Tiengrim a, Pattarachai Kiratisin b, Visanu Thamlikitkul a, * KEYWORDS Colistin;

against Clinical Isolates of Gram-Positive Bacteria

Intrinsic, implied and default resistance

International Journal of Health Sciences and Research ISSN:

What s next in the antibiotic pipeline?

The International Collaborative Conference in Clinical Microbiology & Infectious Diseases

Chapter 51. Clinical Use of Antimicrobial Agents

Polymyxins Revisited

Brief reports. Decreased susceptibility to imipenem among penicillin-resistant Streptococcus pneumoniae

Scottish Medicines Consortium

Tackling the need for new antibacterial drugs

Sepsis is the most common cause of death in

Antimicrobial Stewardship Strategy: Antibiograms

Principles of Anti-Microbial Therapy Assistant Professor Naza M. Ali. Lec 1

SUMMARY OF PRODUCT CHARACTERISTICS. Lincomycin (as Lincomycin hydrochloride) Neomycin (as Neomycin sulphate) Excipients Disodium edetate

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

Introduction to Pharmacokinetics and Pharmacodynamics

ANTIBIOTICS USED FOR RESISTACE BACTERIA. 1. Vancomicin

Isolation of antibiotic producing Actinomycetes from soil of Kathmandu valley and assessment of their antimicrobial activities

Comparison of Antibiotic Resistance and Sensitivity with Reference to Ages of Elders

Antibiotics. Antimicrobial Drugs. Alexander Fleming 10/18/2017

DETERMINANTS OF TARGET NON- ATTAINMENT IN CRITICALLY ILL PATIENTS RECEIVING β-lactams

Mono- versus Bitherapy for Management of HAP/VAP in the ICU

Summary of unmet need guidance and statistical challenges

EDUCATIONAL COMMENTARY - Methicillin-Resistant Staphylococcus aureus: An Update

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)

TEST REPORT. Client: M/s Ion Silver AB. Loddekopinge. Sverige / SWEDEN. Chandran. min and 30 min. 2. E. coli. 1. S. aureus

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

Selective toxicity. Antimicrobial Drugs. Alexander Fleming 10/17/2016

SURVIVABILITY OF HIGH RISK, MULTIRESISTANT BACTERIA ON COTTON TREATED WITH COMMERCIALLY AVAILABLE ANTIMICROBIAL AGENTS

RETROSPECTIVE STUDY OF GRAM NEGATIVE BACILLI ISOLATES AMONG DIFFERENT CLINICAL SAMPLES FROM A DIAGNOSTIC CENTER OF KANPUR

UCSF guideline for management of suspected hospital-acquired or ventilatoracquired pneumonia in adult patients

Challenges Emerging resistance Fewer new drugs MRSA and other resistant pathogens are major problems

A snapshot of polymyxin use around the world South America

Human health impacts of antibiotic use in animal agriculture

Efficacy of Colistin in combination with Carbapenem and Tigecycline in patients with pneumonia caused by multidrug-resistant Acinetobacter baumannii

Antimicrobial resistance (EARS-Net)

Antibiotic Stewardship in Nursing Homes SAM GUREVITZ PHARM D, CGP ASSOCIATE PROFESSOR BUTLER UNIVERSITY COLLEGE OF PHARMACY AND HEALTH SCIENCE

Pharmacokinetic-pharmacodynamic profiling of four antimicrobials against Gram-negative bacteria collected from Shenyang, China

Antibacterial activity of Stephania suberosa extract against methicillin-resistant Staphylococcus aureus

Antimicrobial Stewardship/Statewide Antibiogram. Felicia Matthews Senior Consultant, Pharmacy Specialty BD MedMined Services

Transcription:

Journal of Antimicrobial Chemotherapy Advance Access published February 8, 2007 Journal of Antimicrobial Chemotherapy doi:.93/jac/dkl52 In vitro pharmacodynamics of colistin against Acinetobacter baumannii clinical isolates Roxanne J. Owen, Jian Li *, Roger L. Nation and Denis Spelman 2 Facility for Anti-infective Drug Development and Innovation, Victorian College of Pharmacy, Monash University, 38 Royal Parade, Parkville, Victoria 3052, Australia; 2 Department of Microbiology and Infectious Diseases Unit, Alfred Hospital, Australia Introduction Received 3 July 2006; returned 6 September 2006; revised and accepted 20 November 2006 Background: Colistin is being increasingly used for treatment of infections caused by multidrug-resistant Gram-negative bacteria, including Acinetobacter baumannii. Methods: Thein vitro pharmacodynamic properties of colistin (sulphate) were investigated by studying the time kill kinetics and the post-antibiotic effect (PAE) against multidrug-resistant, including colistin heteroresistant, A. baumannii. Time kill was studied with four multidrug-resistant clinical isolates at concentrations ranging from 0.5 to 643 MIC. The PAE was examined after 20 min exposure with five clinical isolates, including the four in the time kill study, plus ATCC 9606. Results: Colistin showed extremely rapid killing in a concentration-dependent manner; but re-growth was observed as early as 3 h and substantial re-growth at 24 h even at concentrations up to 323 MIC or 643 MIC for some isolates. Colistin exhibited modest PAE of.0, 2.3 and 3.5 h at 6, 32 and 643 MIC, respectively, against ATCC 9606. Surprisingly, negative PAE (range: 0.8 to 8.5 h) was observed for all of the five clinical isolates. Conclusions: These findings suggest that monotherapy with colistin methanesulphonate, the parenteral form of colistin, and long dosage intervals (e.g. 24 h) may be problematic for treatment of infections caused by colistin heteroresistant A. baumannii. Keywords: killing kinetics, heteroresistance, post-antibiotic effect One of the greatest accomplishments of modern medicine has been the development of antibiotics for the treatment of infections that were fatal. However, this has inevitably been followed by the acquisition of resistance towards their antimicrobial activity. Unfortunately, the past two decades have seen a remarkable increase in resistance to the currently available antibiotics and a marked decline in the discovery and development of novel antibiotics.,2 In particular, multidrug-resistant Gram-negative bacteria are a difficult target for new antibiotic development, mainly due to the poor permeability posed by their outer membranes and efflux pumps. Indeed, no new antibiotic classes are expected to be commercially available within the next several years against multidrug-resistant Gram-negative bacteria, including Pseudomonas aeruginosa and Acinetobacter baumannii. It is precisely this scenario to which the IDSA refers in its Bad bugs, no drugs campaign. A. baumannii is an opportunistic Gram-negative pathogen that may cause pneumonia, bacteraemia, infection in burn wounds, meningitis and urinary tract infections. 3 Infections caused by A. baumannii in war-related injuries have been documented. 4 The mortality rates for A. baumannii infections have been reported to be 52% for bacteraemia and 23 73% for pneumonia. 5 Unfortunately, resistance to aminoglycosides, carbapenems, cephalosporins and fluoroquinolones in multidrug-resistant A. baumannii clinical isolates has substantially increased worldwide in the last decade 3,5 7 and it is regarded as one of the most difficult nosocomially-acquired Gram-negative pathogens to treat and control. 2 The recently approved tigecycline is a therapeutic option for adults with complicated skin and skin structure infections caused by multidrug-resistant A. baumannii; however, the potential toxicities similar to tetracycline and its unavailability in many countries have limited its clinical applications. 2 In this setting of increasing resistance and diminishing therapeutic options, the old antibiotic colistin is now being used more extensively. 8... *Corresponding author. Tel: þ6-3-9903-9702; Fax: þ6-3-9903-9629; E-mail: Jian.Li@vcp.monash.edu.au... Page of 5 # The Author 2007. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy. All rights reserved. For Permissions, please e-mail: journals.permissions@oxfordjournals.org

Owen et al. Colistin, also known as polymyxin E, is a multi-component polypeptide antibiotic that was discovered in the 950s. 9 Owing to its significant (currently high) activity against Gram-negative superbugs, including P. aeruginosa and A. baumannii, and uncommon resistance, colistin is now being administered systemically as salvage therapy in patients where none of the other available antibiotics are active against a patient s isolate. 8 With increased use in recent years, it has been possible to more thoroughly assess colistin s potential for causing toxicity; interestingly, it appears to be less nephrotoxic than the aminoglycosides, which were believed to be less toxic and replaced colistin in the 970s. 8 Because colistin was developed before contemporary drug development and approval procedures were introduced, there are substantial gaps in knowledge of its pharmacokinetics, pharmacodynamics and toxicodynamics. 8,,2 Worryingly, resistance to colistin in clinical isolates has been reported recently in Europe. 3,4 Also of very substantial concern is the observation of colistin heteroresistance in 5 out of 6 clinical isolates of colistin-susceptible (colistin MICs 2 mg/l) but multidrug-resistant A. baumannii. 5 Clearly, a comprehensive understanding of its pharmacology is urgently required and will form the basis for strategies to prevent or minimize the development of resistance. The aim of the present study was to examine the in vitro pharmacodynamic properties, namely bacterial killing and the post-antibiotic effect (PAE), of colistin against multidrug-resistant A. baumannii and to compare the magnitude of pharmacodynamic properties with concentrations achieved in vivo. Because sodium colistin methanesulphonate (CMS), the form of colistin for parenteral use, is a non-active pro-drug, 6 only colistin (sulphate) was employed in the current study. Materials and methods Bacterial strains and antibiotics The five A. baumannii clinical isolates, all of which were multidrug-resistant, were obtained from patients at the Alfred Hospital (Melbourne, Australia). The samples were taken from sputum (isolates 3, 6, 7 and ) or bronchoalveolar lavage (isolate 8) between 2 December 2002 and 7 November 2004 and identified with VITEK2 GN card (biomérieux, Australia). A. baumannii ATCC 9606 (VA, USA) was employed as a reference strain. All strains were stored at 808C and subcultured onto horse-blood agar plates (Medium Preparation Unit, University of Melbourne, Australia) before the experiments. Colistin stock solutions were prepared by dissolving powdered colistin sulphate (Lot23K382, Sigma-Aldrich, Castle Hill, NSW, Australia) with Milli-Q water (Millipore, Australia) and sterilizing by passage through 0.20 mm syringe filters (Sartorius, Australia). Time kill kinetics The time killing kinetics of four clinical isolates (isolates 3, 7, 8 and ) by colistin (sulphate, MICs, 0.5, 2 and mg/l, respectively) was examined. The antibiotic was added to a logarithmic-phase broth culture of approximately 6 cfu/ml (5.73 5.9 log cfu/ml) to yield concentrations of 0, 0.5,, 2, 4, 8, 6, 32 and 64 MIC of the respective isolate. Viable counting was performed on samples collected at 0, 20, 40, 60 min and 2, 3, 4, 6 and 24 h after antibiotic addition. After appropriate dilutions with saline, samples of bacterial cell suspension (50 ml) were spirally plated on nutrient agar plates (Medium Preparation Unit) using a Whitley automatic spiral plater (WASP, Don Whitley Scientific, West Yorkshire, UK). Colonies were counted by a ProtoCOL automated colony counter (Synbiosis, Cambridge, UK) after 24 h of incubation of subcultures at 358C. The lower limit of counting was 20 cfu/ml. The killing effect of colistin at different concentrations was quantified by calculation of the mean survival time over 4 h (MST 240 min ). 7 Post-antibiotic effect (PAE) The in vitro PAE was determined by a previously described method 7 for the four clinical isolates noted above, plus isolate 6 and ATCC 9606; the time kill kinetics of the latter two strains had been examined in a recent study. 5 The five clinical isolates and the reference strain belonged to six different PFGE pattern groups. 5 For each PAE experiment, A. baumannii ( 6.5 cfu/ ml) in logarithmic phase growth was exposed for 20 min in CAMHB to colistin (sulphate) at concentrations of 0.5,, 2, 4, 8, 6, 32 and 64 MIC. Exposure of 20 min was used for colistin due to its very rapid bactericidal effect. 5 Antibiotic was removed by centrifuging three times at 3000 g for min, decanting the supernatant and resuspending in pre-warmed broth. Viable counts were performed at 0,, 2, 3, 4, 5, 6, 7, 8, 9,,, 2 and 24 h, onto nutrient agar. A growth control was performed in the same fashion, but without exposure to the antibiotic. The colonies were counted after 24 h of incubation at 358C. PAE was determined by comparing regrowth of treated and growth control cultures, using the standard formula of the time for the control culture to increase -fold subtracted from the time for the treated culture to do the same. 7 Results Time kill kinetics In time kill studies, colistin was bactericidal in a concentrationdependent manner (Figure ); results for isolates 3 and 7 were similar to those for isolates 8 and shown in the figure. At the highest multiples of the MIC, the bacterial killing for all the strains was so rapid that at 64 MIC, no bacteria could be detected (lower limit of counting 20 cfu/ml) within 20 min. Re-growth was observed as early as 3 h and substantial re-growth occurred at 24 h in all clinical isolates at multiples of the respective MICs. For isolate 3, regrowth occurred at 24 h even at 64 MIC, while for isolates 7, 8 and it was observed at concentrations up to 32 MIC, 6 MIC and 4 MIC, respectively (Figure ). Mean survival times (MST 240 min ) were calculated at different multiples of the MIC (Figure 2). Both the abscissa and the ordinate of this figure are plotted on a logarithmic scale to assist in comparisons across strains. The killing kinetics of colistin against ATCC 9606 and isolate 6 were reported previously, 5 and, in the current study, their MST 240 min was calculated and compared with the four clinical isolates reported here. At 0.5 and MIC, all five of the clinical isolates and ATCC 9606 had mean survival times in the range of 82 22 min. At concentrations between 2 and 32 MIC, there were substantial differences among these strains (Figure 2). With the exception Page 2 of 5

Pharmacodynamics of colistin 8 (a) (b) Log cfu/ml 6 4 2 0 0 2 4 6 8 22 24 0 2 4 6 8 22 24 Hours Hours Figure. Time kill curves for colistin against A. baumannii isolates 8 (a) and (b). of ATCC 9606 which had an MST 240 min less than min at 4 MIC and above, the MST 240 min of all the clinical isolates decreased to less than min only at concentrations of at least 6 or 32 MIC. Post-antibiotic effects (PAEs) Figure 3 illustrates the PAEs of colistin on the five clinical isolates and the reference strain. Substantially different profiles were obtained among the strains. Modest PAE was observed for ATCC 9606 (.0 3.5 h) only at concentrations of 6 MIC and above, and for isolates 6 and 8 (0.3 h and.2 h, respectively) at 64 MIC. Surprisingly, substantial negative PAEs Mean survival time (min) 00 0 0. ATCC 9606 Isolate 3 Isolate 6 Isolate 7 Isolate 8 Isolate Multiples of MIC Figure 2. Bactericidal activities of colistin against A. baumannii isolates as measured by MST 240 min. Due to the rapid killing, MST 240 min values could not be calculated for isolate 7 at 6 MIC and above, for ATCC 9606 and isolates 8 and at 32 and above, and for isolates 3 and 6 at 64 MIC. 0 Control 0.5 MIC MIC 2 MIC 4 MIC 8 MIC 6 MIC 32 MIC 64 MIC ranging from 0.8 to 8.5 h were observed for all the clinical isolates at various multiples of MICs (Figure 3). Discussion Many clinicians have already been confronted with the reality of infections with Gram-negative bacteria, including A. baumannii resistant to all the currently available antibiotics except colistin. 8 Unfortunately, emergence of resistance to colistin has been recently reported in multidrug-resistant A. baumannii. 3 Equally worrying, heteroresistance to colistin was discovered recently in multidrug-resistant but colistin-susceptible (based on MICs) A. baumannii. 5 Our knowledge on the pharmacology of colistin is still limited 8 and there has been no systematic investigation on the in vitro pharmacodynamics of colistin against A. baumannii. Inthe Postantibiotic effect (hours) 6 4 2 0 2 4 6 8 ATCC 9606 Isolate 3 Isolate 6 Isolate 7 Isolate 8 Isolate Multiples of MIC Figure 3. PAE of colistin against A. baumannii isolates. 0 Page 3 of 5

Owen et al. current study, the killing kinetics and PAE of colistin were examined against multidrug-resistant A. baumannii clinical isolates. Colistin was very active in the initial killing (Figure ); even with 0.5 MIC, the log cfu/ml decreased at least.5 (with maximum decrease of 4.26 for isolate 3) within 4 h for the four clinical strains tested. Arguably, the most important finding in this study was the substantial regrowth observed at 24 h even at 64 MIC. This is consistent with our recent observation of heteroresistance to colistin in these isolates. 5 The more resistant sub-populations probably led to the substantial regrowth at 24 h. Considering that achieved plasma colistin concentrations are in the range of 4 mg/l after intravenous administration of CMS in humans, 8,9 the concentrations required to inhibit any regrowth at 24 h (64 MIC, 6 28 mg/l) are definitely unachievable in vivo, even without considering plasma protein binding. Therefore, this study strongly suggests that care is needed when CMS is administered parenterally as monotherapy for infections caused by multidrug-resistant A. baumannii. The MST 240 min patterns of the tested isolates are slightly different (Figure 2). For all the tested isolates except number 7, decreases in the MST 240 min were observed at concentrations above or 2 MIC. The MST 240 min of isolate 7 did not decrease until the concentration reached 8 MIC, above which the killing was so rapid that the MST 240 min could not be calculated. The values of MST 240 min at 0.5 MIC for clinical isolates 3, 7, 8 and, plus ATCC 9606 and isolate 6, 5 were in the range of 97 22 min (Figure 2), which were longer than those observed previously for P. aeruginosa (3.7 84 min). 7 In addition, unlike P. aeruginosa in which the MST 240 min of all the tested strains fell below min at colistin concentrations of MIC and higher, 7 the A. baumannii clinical isolates reported here required at least 6 MIC for the MST 240 min to be shorter than min. These differences are likely due to more extensive bacterial regrowth for A. baumannii than P. aeruginosa, resulting from the resistant sub-populations in the former species. Colistin showed only modest positive PAE against ATCC 9606 at relatively high concentrations (6 MIC) which are not achievable clinically. 8,9 In contrast, all the clinical isolates exhibited negative PAEs at the tested concentrations; isolates 3 and 7 showed substantial negative PAE ( 5.30 to 8.5 h, respectively) at 0.5 MIC (Figure 3). Such negative PAEs for colistin were not observed against P. aeruginosa. 7 Negative PAE is not commonly observed with any antibiotic/bacterium combinations; it has been reported with meropenem against P. aeruginosa, Escherichia coli and Klebsiella pneumoniae ( 0.2 to 0.8 h). 20 In imipenem-treated E. coli, negative PAE ( 0. to 3.7 h) was suggested to be due to the lysis of osmotically fragile spheroplasts, which were formed by exposure to imipenem, on agar plates thereby influencing viable counting. 2 A short negative PAE was also observed with the antimicrobial peptide lactoferricin against Staphylococcus aureus (range: 6 to 63 min) and E. coli (range: 3 to 2 min). 22 The mechanism(s) of the colistin-associated negative PAE against A. baumannii clinical isolates is unknown and warrants further investigation. Once-daily administration of CMS in cystic fibrosis patients for infections caused by multidrug-resistant P. aeruginosa may provide satisfactory effectiveness and toxicity profiles. 23 The use of such extended dosage intervals against P. aeruginosa requires a greater understanding of the pharmacokinetic/pharmacodynamic determinants for efficacy and comparative clinical trials against more conventional dosage intervals (8 or 2 h). For infections caused by colistin-heteroresistant A. baumannii, in view of the lack of positive PAE at clinically achievable concentrations of colistin, 8,9 as observed in the current study, and the relatively short half-life (approximately 4 h) of colistin after intravenous CMS, 8,9 it is unlikely that once-daily administration of CMS would be a good option for the treatment of such infections. Further preclinical pharmacodynamic investigation is required. In summary, the current study demonstrated initial concentration-dependent bacterial killing against A. baumannii, which was not surprising given that all of the strains studied were colistin-susceptible on the basis of MIC values. Undoubtedly, the most significant findings were the substantial regrowth that occurred even at colistin concentrations up to 64 MIC and the minor or negative PAE of colistin. These findings send a strong warning that monotherapy with CMS and extended-interval (e.g. 24 h) dosage regimens may be problematic for treatment of infections caused by colistinheteroresistant A. baumannii. Acknowledgements We thank the Infectious Diseases Unit, Alfred Hospital, Australia for technical support in identification of the isolates, and Professor John Turnidge, Women s and Children s Hospital, Adelaide, Australia for helpful discussion. This study was presented in part at the Seventh Annual Meeting of the Australian Society for Antimicrobials, Sydney, Australia, 2006. Disclosure of all financial support: we acknowledge the partial financial support of the Australian National Health and Medical Research Council. Transparency declarations Statement of proprietary interest: we do not have any financial, commercial or proprietary interest in any drug, device or equipment mentioned in this paper. References. Infectious Diseases Society of America. 2004. Bad Bugs, No Drugs. http://www.idsociety.org/pa/idsa_paper4_final_web.pdf (20 November 2006, date last accessed). 2. Talbot GH, Bradley J, Edwards JE et al. Bad bugs need drugs: an update on the development pipeline from the Antimicrobial Availability Task Force of the Infectious Diseases Society of America. Clin Infect Dis 2006; 42: 657 68. 3. Bergogne-Berezin E, Towner KJ. Acinetobacter spp. as nosocomial pathogens: microbiological, clinical, and epidemiological features. Clin Microbiol Rev 996; 9: 48 65. 4. Davis KA, Moran KA, McAllister CK et al. Multidrug-resistant Acinetobacter extremity infections in soldiers. Emerg Infect Dis 2005; : 28 24. 5. Jain R, Danziger LH. Multidrug-resistant Acinetobacter infections: an emerging challenge to clinicians. Ann Pharmacother 2004; 38: 449 59. 6. Livermore DM. The threat from the pink corner. Ann Med 2003; 35: 226 34. 7. Livermore DM. The need for new antibiotics. Clin Microbiol Infect 2004; Suppl. 4: 9. Page 4 of 5

Pharmacodynamics of colistin 8. Li J, Nation RL, Milne RW et al. Evaluation of colistin as an agent against multi-resistant Gram-negative bacteria. Int J Antimicrob Agents 2005; 25: 25. 9. Koyama Y, Kurosasa A, Tsuchiya A et al. A new antibiotic colistin produced by spore-forming soil bacteria. J Antibiot 950; 3: 457 8.. Al-Aloul M, Miller H, Alapati S et al. Renal impairment in cystic fibrosis patients due to repeated intravenous aminoglycoside use. Pediatr Pulmonol 2005; 39: 5 20.. Li J, Nation RL, Turnidge J et al. Colistin: the re-emerging antibiotic for multidrug-resistant Gram-negative bacterial infections. Lancet Infect Dis 2006; 6: 589 60. 2. Li J, Nation RL. Old polymyxins are back, is resistance close? Clin Infect Dis 2006; 43: 663 4. 3. Gilad J, Eskira S, Riesenberg K et al. Emergence of nosocomial colistin-resistant Acinetobacter baumannii. In: Abstracts of the Forty-fifth Annual Interscience Conference on Antimicrobial Agents and Chemotherapy, Washington, DC, 2005. Abstract K-292, p. 333. American Society for Microbiology, Washington, DC, USA. 4. Antoniadou A, Kontopidou FV, Poulakou G et al. Emerging Colimycin resistant strains of Klebsiella pneumoniae from intensive care unit patients. In: Abstracts of the Forty-fifth Annual Interscience Conference on Antimicrobial Agents and Chemotherapy, Washington, DC, 2005. Abstract K-507, p. 342. American Society for Microbiology, Washington, DC, USA. 5. Li J, Rayner CR, Nation RL et al. Hetero-resistance to colistin in multidrug-resistant Acinetobacter baumannii. Antimicrob Agents Chemother 2006; 50: 2946 50. 6. Bergen PJ, Li J, Rayner CR et al. Colistin methanesulfonate is an inactive pro-drug of colistin against Pseudomonas aeruginosa. Antimicrob Agents Chemother 2006; 50: 953 8. 7. Li J, Turnidge J, Milne R et al. In vitro pharmacodynamic properties of colistin and colistin methanesulfonate against Pseudomonas aeruginosa isolates from patients with cystic fibrosis. Antimicrob Agents Chemother 200; 45: 78 5. 8. Li J, Rayner CR, Nation RL et al. Pharmacokinetics of colistin methanesulfonate and colistin in a critically ill patient receiving continuous venovenous hemodiafiltration. Antimicrob Agents Chemother 2005; 49: 484 5. 9. Li J, Coulthard K, Milne R et al. Steady-state pharmacokinetics of intravenous colistin methanesulphonate in patients with cystic fibrosis. J Antimicrob Chemother 2003; 52: 987 92. 20. Hanberger H, Svensson E, Nilsson LE et al. Pharmacodynamic effects of meropenem on gram-negative bacteria. Eur J Clin Microbiol Infect Dis 995; 4: 383 90 2. Hanberger H, Svensson E, Nilsson M et al. Effects of imipenem on Escherichia coli studied using bioluminescence, viable counting and microscopy. J Antimicrob Chemother 993; 3: 245 60. 22. Haukland HH, Vorland LH. Post-antibiotic effect of the antimicrobial peptide lactoferricin on Escherichia coli and Staphylococcus aureus. J Antimicrob Chemother 200; 48: 569 7. 23. Rosenvinge A, Pressler T, Hoiby N. Colistin intravenously is a safe and effective treatment of multidrugresistant microorganisms in cystic fibrosis. J Cyst Fibros 2005; 4: S32. Page 5 of 5