Nontraditional Therapy for Stenotrophomonas maltophilia Infections: Ending the Sulfuring

Similar documents
Tested Against Tigecycline and Agents Commonly Used for S. maltophilia Infections. David J. Farrell 1*, Helio S. Sader 1,2. and. Ronald N.

Tetracycline Rationale for the EUCAST clinical breakpoints, version th November 2009

Most common dose (mg) 1g x 1 1g x 1 1g x 1 1g x 1 1g x 1 1g x 1. Maximum dose schedule (mg) 1g x 1 1g x 1 1g x 1 1g x 1 1g x 1 1g x 1

Antimicrobial Activities of Ceftazidime/Avibactam and Comparator Agents against Clinical. Bacteria Isolated from Patients with Cancer.

Nitroxoline Rationale for the NAK clinical breakpoints, version th October 2013

Antibiotic Resistance in Enterobacteriaceae

EUCAST. Linezolid Rationale for the EUCAST clinical breakpoints, version th November 2005

Antimicrobial Susceptibility of Stenotrophomonas maltophilia Isolates from Korea, and the Activity of Antimicrobial Combinations against the Isolates

Gentamicin Rationale for the EUCAST clinical breakpoints, version th February, 2009

A Combination of Trimethoprimsulfamethoxazole. Showed Good In Vitro Activity against. Original Article. Abstract. Introduction

Stenotrophomonas maltophilia bacteremia in pediatric patients a 10-year analysis

Toronto General Hospital ANTIBIOGRAM Emergency Department January 1, December 31, 2016

Stenotrophomonas maltophilia in cystic fibrosis: Improved detection by the use of selective agar and evaluation of antimicrobial resistance

Analysis of Longitudinal Data. Patrick J. Heagerty PhD Department of Biostatistics University of Washington

b-lactam resistance and b-lactamase expression in clinical Stenotrophomonas maltophilia isolates having defined phylogenetic relationships

A multicenter surveillance of antimicrobial resistance in Serratia marcescens in Taiwan

Validation of EUCAST zone diameter breakpoints against reference broth microdilution

Pseudomonas putida 5

Evaluation of a Stenotrophomonas maltophilia bacteremia cluster in hematopoietic stem cell transplantation recipients using whole genome sequencing

STUDY ABOUT ANTIBIOTIC RESISTANCE IN SERRATIA SPP. ISOLATED FROM HOSPITALIZED PATIENTS

CHAPTER 3 HEART AND LUNG TRANSPLANTATION. Editors: Mr Mohamed Ezani Md. Taib Dato Dr David Chew Soon Ping Dr Ashari Yunus

CHAPTER 3 HEART AND LUNG TRANSPLANTATION. Editors: Mr Mohamed Ezani Md. Taib Dato Dr David Chew Soon Ping Dr Ashari Yunus

Why the CDS? The unique advantages of using an Australian antimicrobial susceptibility testing method

THE IDENTIFICATION OF TWO UNKNOWN BACTERIA AFUA WILLIAMS BIO 3302 TEST TUBE 3 PROF. N. HAQUE 5/14/18

Antimicrobial therapy for Stenotrophomonas maltophilia infections

Risk Assessment of Staphylococcus aureus and Clostridium perfringens in ready to eat Egg Products

EUCAST Expert Rules Version 3.1. Intrinsic Resistance and Exceptional Phenotypes Tables

Most common dose (mg) 1 g x 3 1 g x mg -1.0 g x 3 1 g x mg -1 g x mg -1 g x 3

Expression of Sme Efflux Pumps and Multilocus Sequence Typing in Clinical Isolates of Stenotrophomonas maltophilia

Gram negative bacilli

Department of Microbiology and Clinical Microbiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey

Alita Miller, PhD. Head of BioScience. April 24, 2017 ECCMID 2017 Vienna, Austria

The Genetic Epidemiology of Antibiotic Resistance

Report: antimicrobial resistance in commensal E. coli from poultry, pigs, cows and veal calves. 2014

Antimicrobial Resistance in Haemophilus influenzae

Implementation of Public Health Surveillance of Carbapenemase- Producing Enterobacteriaceae in Victoria, Australia

HAEMOPHILUS MODULE 29.1 INTRODUCTION OBJECTIVES 29.2 MORPHOLOGY. Notes

by author ESCMID Online Lecture Library Epidemiological cutoff values (ECOFFs) and Low Level resistance Gunnar Kahlmeter

Expansion of Salmonella Typhimurium ST34 clone carrying multiple. resistance determinants in China

Antibiotic resistance by efflux: from molecular aspects to cinical impact

Antibiotic Resistance in Escherichia coli Iron Transport Mutants

PHA 4123 First Exam Fall On my honor, I have neither given nor received unauthorized aid in doing this assignment.

Efflux Mechanisms of Fluoroquinolones and β-lactams

Modular Program Report

Wayne State University Doctor of Pharmacy Program. Curriculum and Course Prerequisites Classes of 2017, 2018, and 2019

Study No: Title : Rationale: Phase: Study Period: Study Design: Centres: Indication: Treatment: Objectives: Statistical Methods:

Antibiotic efflux pumps in eucaryotic cells: consequences for activity against intracellular bacteria

In vitro the effect of intestinal normal flora on some pathogenic bacteria.

NCCTG Status Report for Study N May 2010

Sample Date: March 30, 2018 Date Received: March 31, 2018 Date of Report: April 9, 2018 (877) Fax: (877)

Comparative effectiveness of dynamic treatment regimes

μ gyra parc Escherichia coli Klebsiella pneumoniae Pseudomonas aeruginosa E. coli gyra E. coli parc gyra parc gyra Escherichia coli E. coli E.

Research of New Class C beta-lactamase Inhibitor by Molecular Docking s Method

Research Article Evaluation of Verigene Blood Culture Test Systems for Rapid Identification of Positive Blood Cultures

The general concept of pharmacokinetics

Department of Medical Microbiology, Istanbul University Cerrahpas a Medical Faculty, Istanbul, Turkey; 3 SUMMARY

Chronic Granulomatous Disease Medical Management

Available from Deakin Research Online:

Salmonella enteritidis Identification and Isolation

Antibiotic efflux pumps in eucaryotic cells: consequences for activity against intracellular bacteria

Typhoid Fever Dr. KHALID ALJARALLAH

2 Salmonella Typhimurium

Serratia marcescens (

cefixime CFIX cefteram pivoxil CFTM-PI ceftriaxone CTRX cefodizime CDZM spectinomycin

BIOL 51A - Biostatistics 1 1. Lecture 1: Intro to Biostatistics. Smoking: hazardous? FEV (l) Smoke

Distributed analysis in multi-center studies

PubH 7470: STATISTICS FOR TRANSLATIONAL & CLINICAL RESEARCH

Supplementary Appendix

by author What is new in EUCAST?

Multidrug resistant Stenotrophomonas maltophilia: an emerging cause of hospital acquired infections in Assiut University Hospitals, Egypt

Effect of iron and sodium chloride on biofilm development of stenotrophomonas maltophilia

PubH 7405: REGRESSION ANALYSIS MLR: BIOMEDICAL APPLICATIONS

Three Steps to Antibiotic Resistance?

This presentation contains forward-looking statements within the meaning of the "safe harbor" provisions of the Private Securities Litigation Reform

EASTERN ARIZONA COLLEGE Microbiology

Considerations with Antibiotic Therapy PART

Effluxpumpeninhibitoren alternative Therapiestrategie bei mikrobieller Multiresistenz

Characterization of Class 1 Integrons and Antimicrobial Resistance in CTX-M-3-Producing Serratia marcescens Isolates from Southern Taiwan

Ian Morrissey, 1 Samuel K. Bouchillon, 2 Meredith Hackel, 2 Douglas J. Biedenbach, 2 Stephen Hawser, 1 Daryl Hoban 2 and Robert E.

Increasing Carbapenem-Resistant Gram-Negative Bacilli and Decreasing Metallo-β-Lactamase Producers over Eight Years from Korea

INTRODUCTION MATERIALS & METHODS

ANTIBACTERIAL GRAPHENE OXIDE AND METAL PARTICLES NANOCOMPOSITES FOR INHIBITION OF PATHOGENIC BACTERIA STRAINS

An outbreak of multidrug-resistant Serratia marcescens: The importance of continuous monitoring of nosocomial infections

Bias in Markov Models of Disease

Dial Complete Foaming Antimicrobial Hand Soap

International Journal of Pharma and Bio Sciences

CTP-656 Tablet Confirmed Superiority Of Pharmacokinetic Profile Relative To Kalydeco in Phase I Clinical Studies

CLASSIFICATION OF BACTERIA

The Clinical Aspects of β-lactam-resistant Stenotrophomonas

3 S. Heidelberg ESBL Extended spectrum lactamase

Comparison of cefotiam and cefazolin activity against Gram-negative bacilli

Overview of the major bacterial pathogens The major bacterial pathogens are presented in this table:

Nonlinear pharmacokinetics

Ph.D. course: Regression models. Introduction. 19 April 2012

Modular Program Report

Key words: Staphylococci, Classification, Antibiotic-susceptibility, Opportunistic infection

Stability. Received for publication 1 August to be fl-lactamase-producing strains.

Salmonella empyema: a case report

Hypothesis Testing, Power, Sample Size and Confidence Intervals (Part 2)

Objectives. Epidemiology of AB resistance in S. pneumoniae. Pasteur Institute (R. Vanhoof) AB resistance evaluation (MIC) & mechanisms

Transcription:

Nontraditional Therapy for Stenotrophomonas maltophilia Infections: Ending the Sulfuring Reese Cosimi, Pharm.D., BCPS PGY2 Infectious Diseases Pharmacy Resident South Texas Veterans Health Care System The University of Texas at Austin College of Pharmacy The University of Texas Health Science Center at San Antonio December 9, 2016 Objectives 1. Recognize risk factors and clinical characteristics associated with Stenotrophomonas maltophilia infection 2. Explain mechanisms of Stenotrophomonas maltophilia antimicrobial resistance and its resultant limited susceptibility to traditional agents 3. Recommend optimal antimicrobial therapy for Stenotrophomonas maltophilia in a patient who is not a candidate for trimethoprim-sulfamethoxazole Cosimi 1

Background I. History of Stenotrophomonas maltophilia 1 A. Initially isolated from pleural fluid B. Taxonomically reclassified on multiple occasions Figure 1: Nomenclature 1,2 1943 Bacterium bookeri 1960 Pseudomonas maltophilia 1981 Xanthomonas maltophilia 1993 Stenotrophomonas maltophilia II. Epidemiology A. Incidence 1. Infections due to S. maltophilia occur in 7.4 to 37.7 patients in 10,000 at risk for opportunistic infections 3 2. Isolated in 30% of cystic fibrosis patient sputum cultures 4 B. Mortality 1. Difficult to determine true mortality for several reasons 1 a. Low pathogenicity makes infection versus colonization unclear b. Isolation of S. maltophilia most commonly occurs in critically ill patients c. Poor association with infection and worse clinical outcomes 2. Overall mortality from acute respiratory tract infections was found to be 20.3% though multiple confounders were present 5 a. No evidence of lung consolidation in 70% of patients b. Administration of an antibiotic against S. maltophilia had no effect on outcomes c. Concomitant nosocomial pathogens were present in >30% of patients 3. Attributable mortality with bacteremia is 26.7% and treatment of S. maltophilia improved survival in this population 6 C. Common sources of S. maltophilia 1 1. Soil and plant roots 2. Contaminated fluids a. Dialysate b. Contact solution c. Chlorhexidine d. Sink drains and handwashing soap e. Ice machines Cosimi 2

D. Patients at highest risk for isolation of S. maltophilia are those with multiple comorbidities Table 1: Risk Factors 1,6 Malignancy Indwelling devices Chronic respiratory disease Immunocompromised host Prior antibiotic use Prolonged hospitalization ICU admission Chronic conditions E. Typical infectious presentation 1,7 1. Majority of infections are polymicrobial 2. Pneumonia, bacteremia and catheter-related blood stream infections are most common 3. COPD and cystic fibrosis exacerbations, skin and soft tissue infections, osteomyelitis, meningitis, and intraabdominal infections are also seen III. Microbiology 1 A. Yellow colony cultures B. Gram-negative bacilli with polar flagella C. Non-lactose fermenter D. Obligate aerobe E. Oxidase negative F. High affinity to form biofilm on plastic devices Figure 3: Biofilm Formation 9 Cosimi 3

G. Clinical and Laboratory Standards Institute (CLSI) breakpoints Table 2: Breakpoints 10 Drug Susceptible Intermediate Resistant TMP-SXT 2/38 --- 4/76 Minocycline 4 8 16 Levofloxacin 2 4 8 Ceftazidime 8 16 32 TMP-SMX = trimethoprim-sulfamethoxazole IV. Pharmacotherapy A. High rates of intrinsic resistance makes antibiotic selection difficult 11 1. TMP-SMX is considered treatment of choice 12 2. Historical data supporting alternative/combination therapy was lacking B. S. maltophilia hospital-acquired pneumonia or ventilator-associated pneumonia 1. Infectious Diseases Society of America (IDSA) makes no recommendations 13 2. Original in vitro studies demonstrated highest susceptibility with TMP-SMX which supported its utility 11,14,15 C. S. maltophilia catheter-related blood stream infections 1. IDSA guidelines recommend removal of infected device plus TMP-SMX administration 16 2. Recommendation based on original high susceptibility of TMP-SMX plus evidence that receipt of appropriate therapy decreased mortality by up to 45% 11,17 D. Clinical data using TMP-SMX is lacking 1. 86 ventilator-associated pneumonia patients = 13% mortality 18 2. 18 bacteremia patients = 0% mortality 19 3. 26 patients with all infection types = 30.8% mortality 20 V. Pharmacology of agents active against S. maltophilia Table 3: Pharmacology 2 TMP-SMX 16,22 Minocycline 23 Levofloxacin 24 Dose 3-5mg/kg every 8 100mg twice 750mg daily hours daily Route IV or oral IV or oral IV or oral Distribution Vd (L/kg) Cmax (mg/l) Protein Bound (%) TMP: 2; SMX: 0.36 TMP: 3.4; SMX: 46.3 TMP: 44; SMX: 70 1.17 4.1 76 1.1 0.6-9.4 38 Metabolism Hepatic Hepatic None Renal Elimination (%) TMP: 37-85 5-12 80 Adverse Drug Reactions SMX: 17-67 Hypersensitivity Nephrotoxicity Hemolytic anemia Thrombocytopenia Hyperkalemia GI upset Hepatotoxicity Photosensitivity Vd = Volume of distribution; CDI = Clostridium difficile infection Tendon rupture CDI Neurotoxicity Prolonged QT Cosimi 4

VI. In vitro susceptibility and resistance mechanisms A. Gram-negative pathogens often have multiple intrinsic and acquired mechanisms of resistance 25 Figure 4: Mechanisms of Resistance 26 Figure 4: Mechanisms of Resistance 26 Fluoroquinolones Tetracyclines Beta-lactams Aminoglycoside B. S. maltophilia is resistant to many antimicrobials commonly used for gram-negative infections 27 1. TMP-SMX 28 a. Mechanism: acquired mobile sul-genes increase bacterial fitness against TMP-SMX b. Increasing prevalence noted worldwide 2. Beta-lactams a. Mechanism: two types of inducible beta-lactamases i. Metallo-beta-lactamase (L1) ii. Clavulanate sensitive cephalosporinase (L2) b. Resistance: All penicillins, cephalosporins and carbapenems c. Ceftazidime and ticarcillin-clavulanate may retain susceptibility though low clinical cure rates have been reported (50 and 60%, respectively) 3. Aminoglycosides a. Mechanism: enzymatic inactivation via acetyltransferase b. Resistance: All aminoglycosides 4. Fluoroquinolones 29 a. Mechanism: efflux pumps and decreased uptake b. Resistance: All fluoroquinolones c. Only levofloxacin has susceptibility breakpoints d. In vitro moxifloxacin is most susceptible but may not equate in vivo 5. Tetracyclines 30 a. Mechanism: efflux pumps and decreased uptake b. Resistance: All tetracyclines Cosimi 5

C. Previous antimicrobial exposure significantly increases risk for expression of resistance 11 Table 4: Recent Antibiotic Susceptibility Drug Percent Susceptible (%) Minocycline 33-35 95 100 TMP-SMX 12,31,33-34,37-38 80 100 Levofloxacin 31-35,37-38 44 95 Tigecycline 34-35 80 84 Moxifloxacin 12,34-35 75 80 Ticarcillin-clavulanate 12,34,37 41 76 Piperacillin 32 57 Ceftazidime 12,31-34,37-38 11 39 Colistin 34 23 Ciprofloxacin 36 22 Gentamicin 32 2 *Antimicrobials without breakpoints required extrapolation from other organisms or drugs in many of these studies VII. Why use alternative agents? A. Hypersensitivity in up to 2% of patients 39 B. Adverse drug reactions in up to 13% of cases 40 C. Thrice daily dosing increases workload and volume burden 21 D. IV formulation drug shortages may take place (most recent: 2014) 41 E. Contraindications to therapy 21 1. History of sulfonamide- or TMP-induced thrombocytopenia 2. Age <2 months 3. Hepatic damage 4. Megaloblastic anemia due to folate insufficiency 5. Renal insufficiency F. Increasing resistance seen in 30-40% of isolates worldwide 11,28 VIII. Which alternative agent? A. Minocycline and levofloxacin are most attractive alternatives to TMP-SMX 1. High susceptibility rates 2. More favorable adverse drug reaction profiles 3. In vitro analyses demonstrate efficacy IX. Clinical Question A. Do clinical data support non-tmp-smx based therapy as an alternative to TMP-SMX? B. Which situations may it be utilized? Cosimi 6

Literature Review Wang YL, Scipione MR, Dubrovskaya Y, Papadopoulos J. Monotherapy with fluoroquinolone or trimethoprim-sulfamethoxazole for treatment of Stenotrophomonas maltophilia infections. Antimicrob Agents Chemother. 2014;58(1):176-182. 42 Objective Assess efficacy of fluoroquinolone monotherapy vs. TMP-SMX in S. maltophilia infections Methods Study Design Retrospective review of patients treated with either a fluoroquinolone (ciprofloxacin or levofloxacin) or TMP-SMX for S. maltophilia infections Location: NYU Langone Medical Center, New York, NY Susceptibility breakpoint source: not reported Population Inclusion Criteria Exclusion Criteria 18 years Received combination therapy Positive S. maltophilia culture Clinical signs/symptoms of nosocomial infection Received S. maltophilia treatment for 48 hours Endpoints Microbiological cure and clinical response at end of therapy (EOT), in-hospital mortality, 30-day mortality and nonsusceptible isolates within 30 days of EOT Microbiological cure definition: no growth from same site at or prior to EOT Statistical Categorical data analyzed with chi-squared or Fisher s exact test Analyses Continuous data analyzed with Student s T test or Mann-Whitney U test P-value of <0.05 indicated statistical significance Multivariate logistic regression Results Enrollment 98 patients were included in the review including 35 patients on TMP-SMX and Baseline Characteristics 63 patients on a fluoroquinolone (48 levofloxacin; 15 ciprofloxacin) Characteristic Fluoroquinolone TMP-SMX (n=63) (n=35) Age (mean ± SD) 69±15 73±15 -- Most common underlying conditions, n(%) Major surgery Malignancy (solid organ) Coronary Artery Disease Diabetes mellitus Mechanical ventilation Intra-abdominal drain Most common sites of infection, n(%) Pulmonary Skin/skin structure Urine Intra-abdominal Bacteremia 28 (44) 24 (38) 26 (41) 26 (41) 17 (27) 1 (2) 38 (60) 9 (14) 6 (10) 5 (8) 5 (8) 14 (40) 14 (40) 11 (31) 9 (26) 12 (34) 6 (17) 17 (49) 10 (29) 3 (9) 4 (11) 1 (3) P-value 0.67 0.85 0.34 0.12 0.45 <0.01 0.26 0.09 1 0.72 0.42 Prior antibiotic use, n(%) 54 (86) 28 (80) 0.46 ICU Admission, n(%) 17 (27) 6 (17) 0.27 Cosimi 7

Length of stay prior to culture, days, median (IQR) Days to treatment, days, median (IQR) Polymicrobial infection, n(%) 53 (84) Pseudmonas spp. 8 (28) Enterococcus spp. 10 (40) Methicillin-resistant 11 (44) Staphylococcus aureus IQR = Interquartile range Results Outcome Fluoroquinolone (n=63) Microbiological cure at 6 (1-19) 4 (0-11) 0.79 3 (2-4) 3 (2-5) 0.98 22 (63) 2 (22) 5 (38) 3 (31) 0.02 0.49 0.83 0.43 TMP-SMX P-value (n=35) 23/37 (62%) 13/20 (65%) 0.83 EOT, n(%) In-hospital mortality, (n%) 16/63 (25%) 7/35 (20%) 0.55 30-day mortality, n(%) 16/52 (31%) 6/27 (22%) 0.42 Clinical success at EOT, n(%) 27/52 (52%) 17/28 (61%) 0.45 Non-susceptible isolate within 30 11/37 (30%) 4/20 (20%) 0.43 days of EOT, n(%) Duration of therapy, days, median 9 (2-38) 8 (2-28) 0.265 (IQR) Length of stay, days, median (IQR) 25 (15-37) 16 (8-42) 0.97 Multivariate analysis identified risk factors for increased mortality: o ICU admission (odds ratio, 8.2; 95% confidence interval, 1.9-35; p = 0.005) o Recent chemotherapy administration (odds ratio, 6.2; 95% confidence interval, 1.12-34; p = 0.037) High rates of resistance were reported after prior exposure to antibiotics in patients with follow-up cultures o TMP-SMX exposure resulted in 2/7 (24%) resistance to TMP-SMX and 2/5 (40%) resistance to levofloxacin o Fluoroquinolone exposure resulted in 0/11 (0%) resistance to TMP-SMX 10/14 (71%) resistance to levofloxacin and Author s When an alternative to TMP-SMX is required, fluoroquinolones may be used an Conclusions alternative option Discussion Critique Strengths Limitations Take-home Points Combination therapy was an exclusion to the study Follow-up susceptibility testing conducted Adjusted for confounding factors Small retrospective analysis Low proportion of patients could be evaluated for all endpoints Minimal bacteremia patients included in study Resistant isolates excluded No differentiation between colonization and infection Fluoroquinolones may be as efficacious as TMP-SMX in initial treatment of S. maltophilia infections Resistance may develop on therapy but may be more common with fluoroquinolones Cosimi 8

Cho SY, Kang C, Kim J, et al. Can levofloxacin be a useful alternative to trimethoprimsulfamethoxazole for treating Stenotrophomonas maltophilia bacteremia? Antimicrob Agents Chemother. 2014;58(1):581-583. 44 Objective Compare clinical outcomes of TMP-SMX with levofloxacin against S. maltophilia bacteremia Methods Study Design Retrospective review evaluating outcomes of adult patients receiving either TMP-SMX or levofloxacin for S. maltophilia bacteremia Location: Samsung Medical Center, Seoul, South Korea Susceptibility breakpoint source: CLSI Population Inclusion Criteria Exclusion Criteria 18 years Received either IV TMP-SMX or levofloxacin for S. maltophilia bacteremia for >48 hours Isolate resistant to prescribed therapeutic agent Endpoints Primary objective: Overall 30-day mortality Secondary objectives: Length of stay, adverse events and bacteremia recurrence Statistical Analyses Categorical variables: Fisher s exact test and chi-squared Continuous variables: Student s t test Statistical significance defined as p value <0.05 Multivariate logistic regression utilized to evaluate effects of therapy with levofloxacin Results Enrollment Of 203 patients identified with S. maltophilia 86 were included in the study. TMP-SMX was administered to 51 patients and levofloxacin to 35 patients Rates of susceptibility to initial S. maltophilia isolates were 96.1% for TMP- SMX and 87.1% for levofloxacin Baseline Characteristics Characteristic TMP-SMX Levofloxacin P value (n=51) (n=35) Age, median (IQR) 56 (40-67) 61 (49-68) 0.153 Gender, male, n(%) 28 (54.9) 28 (80) 0.016 Source of bacteremia, n(%) Definite catheter-related Possible catheter-related Intrabdominal Pneumonia Underlying disease, n(%) Hematologic malignancy Solid tumor Liver cirrhosis Diabetes mellitus Cardiovascular disease Comorbid conditions, n(%) Central venous catheter Chemotherapy Neutropenia 12 (23.5) 11 (21.6) 14 (27.5) 9 (17.6) 29 (56.9) 16 (31.4) 9 (17.6) 8 (15.7) 8 (15.7) 5 (14.3) 11 (31.4) 11 (31.4) 5 (14.3) 15 (44.1) 16 (45.7) 5 (14.3) 5 (14.3) 8 (22.9) 0.29 0.303 0.69 0.678 0.249 0.176 0.678 0.859 0.401 39 (76.5) 30 (58.8) 25 (49) 22 (62.9) 17 (48.6) 13 (37.1) 0.172 0.348 0.276 Catheter removed, n(%) 21 (53.8) 11 (50) 0.773 Cosimi 9

Charlson Comorbidity Index, 2 (2-3) 2 (2-3) 0.539 median (IQR) Septic shock 13 (25.5) 7 (20) 0.554 Polymicrobial bacteremia 12 (23.5) 5 (14.3) 0.290 Combination therapy 9 (17.6) 3 (8.6) 0.345 Typical daily dose 15-20mg/kg 750mg -- Results Outcome TMP-SMX Levofloxacin P value (n=51) (n=35) 30-day mortality, n(%) 14 (27.5) 7 (20) 0.429 Length of stay, days, median 23 (12-51) 27 (15-52) 0.824 (IQR) Adverse events, n(%) 12 (23.5) 0 (0) 0.001 Recurrent bacteremia, n (%) 6 (11.9) 2 (5.7) 0.464 Author s Conclusions No difference noted in 30-day mortality with TMP-SMX versus levofloxacin upon exclusion of patients with combination therapy or polymicrobial infections (29% versus 18.5%; p=0.351) Multivariate analysis identified risk factors associated with increased mortality: o Septic shock (odds ratio, 6.19; 95% confidence interval, 1.81-21.1; p = 0.004) o Pneumonia (odds ratio, 5.33; 95% confidence interval, 1.39-20.37; p = 0.015) Most common adverse events with TMP-SMX reported were rash, cytopenia and hepatotoxicity Half of the recurrent bacteremia isolates were notably resistant to levofloxacin Though clinical trials are needed, levofloxacin may be useful in treating S. maltophilia infections Discussion Critique Strengths Limitations Cohort compared standard of care with levofloxacin Follow-up susceptibility testing conducted Regression analysis conducted to adjust for confounders Only evaluated bacteremia patients Small retrospective analysis Resistant isolates excluded No differentiation between colonization and infection 30-day mortality may not correlate with infection outcomes Take-home Points Levofloxacin may be used for S. maltophilia bacteremia Susceptibility should be determined prior to use as levofloxacin resistance is more common than TMP-SMX Cosimi 10

Jacobson S, Noa LJ, Wallace MR, Bowman MC. Clinical outcomes using minocycline for Stenotrophomonas maltophilia infections. J Antimicrob Chemother. 2016. DOI:10.1093/jac/dkw327. 45 Objective Evaluate efficacy of minocycline for S. maltophilia infections Methods Study Design Retrospective review of patients receiving minocycline for S. maltophilia infections Location: Orlando Regional Medical Center / Orlando Health, Orlando, FL Susceptibility breakpoint source: CLSI Population Inclusion Criteria Exclusion Criteria 18 years Treated with minocycline monoor combination therapy for S. maltophilia Endpoints Clinical failure: Statistical Analyses o o <48 hours of minocycline therapy Pregnant women In-hospital death within 30 days of initial positive culture Clinical deterioration plus one or more of the following: Isolation of S. maltophilia from the same site at follow-up Receipt of an additional antibiotic with in vitro activity against S. maltophilia Partial clinical success: o Lack of clinical deterioration plus one or more of the following: Isolation of S. maltophilia from the same site at follow-up Receipt of additional antibiotics with activity for S. maltophilia Complete clinical success: none of the above Descriptive statistics Results Enrollment 93 of 119 identified patients receiving minocycline for S. maltophilia were included in the review Baseline Characteristics Characteristic Cohort (n=93) ICU, n(%) 49 (53) Mean APACHE II Score, mean (SD) 15 (±6.6) Initial minocycline MIC 4mg/L, n(%) 4mg/L <4mg/L 93 (100) 7 (7.5) 86 (92.5) Infection Type, n(%) Pneumonia Bacteremia 59 (63) 14 (15) Initial Dose 100mg every 12 hours (1 patient received higher dose) Results Outcome Cohort (n=93) Clinical Failure, n(%) 17 (18) Partial Clinical Success, n(%) 22 (24) Complete Clinical Success, n(%) 54 (58) Cosimi 11

Patients more likely to fail therapy: o S. maltophilia minocycline MIC 4mg/L (29.4% vs. 2.6%, p=0.004) o Higher mean APACHE II score (18.1 vs. 14.3, p<0.05) Four patients (9%) on minocycline monotherapy reported clinical failure o 3 deaths; 1 had persistently positive cultures with clinical deterioration o All 4 patients had infections with other microorganisms Author s Minocycline may be an appropriate alternative agent for the treatment of S. Conclusions maltophilia but randomized clinical trials are needed Discussion Critique Strengths Limitations Results supported by existing Retrospective analysis literature Included patients on combination Susceptibility testing conducted therapy without specifying agents for correlation with clinical Concomitant infections were outcomes Patients evaluated for partial common in patients with clinical failure clinical success No differentiation between colonization and infection 30-day mortality may not Take-home Points correlate with infection outcomes Minocycline seems to be effective for the treatment of S. maltophilia infections Less clinically ill patients with lower minocycline MICs may benefit most from minocycline therapy Hand E, Davis H, Kim T, Duhon B. Monotherapy with minocycline or trimethoprim/sulfamethoxazole for treatment of Stenotrophomonas maltophilia infections. J Antimicrob Chemother. 2016;71:1071-1075. 12 Objective Evaluate efficacy of minocycline monotherapy vs. TMP-SMX in the treatment of S. maltophilia infections Methods Study Design Retrospective review of patients receiving either minocycline or TMP-SMX for S. maltophilia infections Location: University Hospital, San Antonio, TX Susceptibility breakpoint source: CLSI Population Inclusion Criteria Exclusion Criteria 1 positive culture for S. maltophilia Received 48 hours of minocycline or TMP-SMX as monotherapy Pregnant Incarcerated Received concominant antibiotics with in vitro activity against S. maltophilia during first 48 hours Endpoints Rates of treatment failure in patients with S. maltophilia treated with minocycline vs. TMP-SMX monotherapy Treatment failure definition: Cosimi 12

o Isolation of S. maltophilia from same site within 30 days o In-hospital death within 30 days of initial S. maltophilia isolate o Receipt of concomitant antibiotic with in vitro activity against S. maltophilia Statistical Demographics and outcomes: descriptive statistics Analyses Continuous data: Mann-Whitney U-test Nominal data: Chi-squared test Logistic regression analyses conducted to account for confounders P-value of <0.05 indicated statistical significance Results Enrollment 45 patients were included in the review including 23 patients on minocycline and 22 patients on TMP-SMX Baseline Characteristic Minocycline TMP-SMX P-value Characteristics (n=23) (n=22) Age, years, mean (range) 54 (18-81) 49 (8-84) 0.42 Most common comorbid condition, n(%) Mechanical ventilation Chronic lung disease Recent ARI Immunosuppression Malignancy Solid Organ Transplant Most common culture site, n(%) Sputum Broncheoalveolar lavage Blood Leukocytosis upon culture isolation, n(%) 13 (57) 12 (52) 10 (43.5) 8 (35) 8 (35) 6 (26) 9 (39) 7 (30) 1 (5) 10 (45) 2 (9) 2 (9) 3 (14) 3 (14) 2 (9) 8 (36) 5 (23) 1 (5) 0.77 0.003 0.017 0.17 0.17 0.24 -- -- -- 12 (52) 12 (55) -- Fever upon culture isolation, n(%) 7 (30) 12 (55) -- ICU Admission, n(%) 17 (74) 14 (64) -- Polymicrobial infections, n(%) P. aeruginosa S. aureus 15 (64) 8 (34) 3 (13) 18 (82) 7 (32) 4 (18) 0.31 -- -- Average daily dose 200mg 8.5mg/kg -- Susceptibility to treatment 23 (100) 22 (100) -- regimen at initiation, n(%) Results Outcome Minocycline TMP-SMX P-value (n=23) (n=22) Treatment failure, n(%) 7 (30) 9 (41) 0.67 S. maltophilia isolated at followup, 5/10 (50) 7/11 (63) 0.75 n(%) Length of stay, days, median 41 (6-136) 54 4-265) 0.35 (range) 30-day in-hospital mortality, n(%) 2 (8.7) 2 (9) -- Duration of therapy, days, median (range) 13 (4-32) 7 (3-15) 0.009 Cosimi 13

No development of resistance to minocycline was noted upon follow-up cultures and susceptibilities Author s Based on the findings of this study, it is reasonable to use minocycline for S. Conclusions maltophilia infections Discussion Critique Strengths Limitations Evaluated solely monotherapy Small retrospective review with patients inadequate power Cohort compared minocycline to Concomitant infections were standard of care common Adjusted for baseline differences Minimal bacteremia patients Follow-up susceptibility testing conducted Attempted to assess colonization versus infection Take-home Points Minocycline has similar clinical success compared with TMP-SMX for the treatment of S. maltophilia infections and may be used as first-line or alternative therapy for this indication Cosimi 14

Conclusions I. Summary A. Evidence continues to grow supporting use of minocycline or fluoroquinolones in the treatment of S. maltophilia B. Levofloxacin is the preferred fluoroquinolone for S. maltophilia based on the presence of CLSI breakpoints and existing clinical data C. Nontraditional therapies may provide equal efficacy as TMP-SMX with more convenient dosing regimens and improved safety profiles II. Recommendations A. TMP-SMX should remain primary therapy for S. maltophilia infections at this time though minocycline susceptibility data supports its use empirically B. When TMP-SMX is not clinically suitable due to contraindications, intolerance, resistant isolates or volume overload, minocycline should be used preferentially to levofloxacin in the treatment of S. maltophilia infections due to the following: 1. Higher rates of susceptibility 2. Better barrier to resistance 3. Superior safety profile 4. Theoretical pharmacokinetic advantages Cosimi 15

References 1. Brooke JS. Stenotrophomonas maltophilia: an emerging global opportunistic pathogen. Clin Microbiol Rev. 2012;25(1):2-41. 2. Denton M, Kerr KG. Microbiological and clinical aspects of infection associated with Stenotrophomonas maltophilia. Clin Microbiol Rev. 1998;11(1):57-80. 3. Garazi M, Singer C, Tai J, Ginochhio C. Bloodstream infections caused by Stenotrophomonas maltophilia: a seven-year review. J Hosp Infect. 2012;81(2):114-118. 4. Davies JC, Rubin BK. Emerging and unusual gram-negative infections in cystic fibrosis. Semin Respir Crit Care Med. 2007;28(3):312-321. 5. Pathmanathan A, Waterer GW. Significance of positive Stenotrophomonas maltophilia culture in acute respiratory tract infection. Eur Respir J. 2005;25:911-914. 6. Senol E, DesJardin J, Stark P, Barefoot L, Snydman D. Attributable mortality of Stenotrophomonas maltophilia bacteremia. Clin Infect Dis. 2002;34(12):1653-1656. 7. Al-Anazi KA, Al-Jasser, AM. Infections caused by Stenotrophomonas maltophilia in recipients of hematopoietic stem cell transplantation. Front Oncol. 2014;4:232. 8. Stenotrophomonas maltophilia. Microregistrar. Updated: August 19, 2014. Accessed: November 11, 2016. http://www.microregistrar.com/tag/stenotrophomonas-maltophilia/ 9. Center for biofilm engineering: biofilm basics. Montana State University. Accessed: November 11, 2016. http://www.biofilm.montana.edu/node/2390 10. CLSI. Performance Standards for Antimicrobial Susceptibility Testing: Twenty-Fifth Informational Supplement. CLSI document M100-S25. Wayne, PA: Clinical Laboratory Standards Institute; 2015. 11. Safdar A. (2015). Stenotrophomonas maltophilia and Burkholderia cepacia. In Bennett JE, Dolin R, Blaser MJ, Mandell, Douglas, and Bennette s Principles and Practice of Infectious Diseases, Updated Edition (pp.2532-2540). Philadelphia: Churchill Livingstone. 12. Hand E, Davis H, Kim T, Duhon B. Monotherapy with minocycline or trimethoprim/sulfamethoxazole for treatment of Stenotrophomonas maltophilia infections. J Antimicrob Chemother. 2016;71:1071-1075. 13. Kalil AC, Metersky ML, Klompas M, et al. Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 clinical practice guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis. 2016;63(5):61-111. 14. Fedler KA, Biedenbach DJ, Jones RN. 2006. Assessment of pathogen frequency and resistance patterns among pediatric patient isolates: report from the 2004 SENTRY Antimicrobial Surveillance Program on 3 continents. Diagn Microbiol Infect Dis. 56:427-436. 15. Al Johani SM, Akhter J, Balkhy H, et al. Prevalence of antimicrobial resistance among gramnegative isolates in an adult intensive care unit at a tertiary center in Saudi Arabia. Ann Saudi Med. 2010;30(5):364-369. 16. Mermel LA, Allon M, Bouza E, et al. Clinical practice guidelines for the diagnosis and management of intravascular catheter-related infections: 2009 update by the Infectious Diseases Society of America. Clin Infect Dis. 2009;49:1-45. 17. Metan G, Uzun O. Impact of initial antimicrobial therapy in patients with bloodstream infections caused by Stenotrophomonas maltophilia. Antimicrob Agents Chemother. 2005;49(9):3980-3981. 18. Czosnowski QA, Wood GC, Magnotti LJ, et al. Clinical and microbiologic outcomes in trauma patients treated for Stenotrophomonas maltophilia ventilator-associated pneumonia. Pharmacotherapy. 2011;31(4):338-345. 19. Lakatos B, Jakopp B, Widmer A, et al. Evaluation of treatment outcomes for Stenotrophomonas maltophilia bacteraemia. Infection. 2014;42(3):553-558. 20. Tekce YT, Erbay A, Cabadak H, Sen S. Tigecycline as a therapeutic option in Stenotrophomonas maltophilia infections. J Chemother. 2012;24(3):150-154. Cosimi 16

21. Micromedex Solutions. Truven Health Analytics, Inc. Ann Arbor, MI. Available at: http://www.micromedexsolutions.com. Accessed November 8, 2016. 22. Sulfamethoxazole and trimethorpime (sulfamethoxazole/trimethoprim) drug summary. 23. Aqwuh KN, MacGowan A. Pharmacokinetics and pharmacodynamics of tetracyclines including glycylcyclines. J Antimicrob Chemother. 2006;58(2);256-265. 24. Fish DN, Chow AT. The clinical pharmacokinetics of levofloxacin. Clin Pharmacokinet. 1997;32(2):101-119. 25. Ruppé E, Woerther PL, Barbier F. Mechanisms of antimicrobial resistance in gram-negative bacilli. Ann Intensive Care. 2015;5:21. 26. Resistance mechanisms in bacteria. ReAct. Accessed: November 8, 2016. http://www.react group.org/toolbox/category /understand/the-rise-and-spread-of-antibiotic-resistance/resistancemechanisms-in-bacteria/. 27. Falagas ME, Valkimadi PE, Huang YT, Matthaiou DK, Hsueh PR. Therapeutic options for Stenotrophomonas maltophilia infections beyond co-trimoxazole: a systematic review. J Antimicrob Chemother. 2008;62(5):889-894. 28. Toleman MA, Bennett PM, Bennett DC, Jones RN, Walsh TR. Global emergence of trimethoprim/sulfamethoxazole resistance in Stenotrophomonas maltophilia mediated by acquisition of sulgenes. Emerg Infect Dis. 2007;13(4)559-565. 29. Grillon A, Schramm F, Kleinberg M, Jehl F. Comparative activity of ciprofloxacin, levofloxacin and moxifloxacin against Klebsiella pneumoniae, Pseudomonas aeruginosa and Stenotrophomonas maltophilia assessed by minimum inhibitory concentrations and time-kill studies. PLoS One. 2016;11(6):1-10. 30. Alonso A, Martinez J. Multiple antibiotic resistance in Stenotrophomonas maltophilia. Antimicrob Agents Chemother. 1997;41(5):1140-1142. 31. Cikman A, et al. Antibiotics resistance of Stenotrophomonas maltophilia strains isolated from various clinical specimens. Afr Health Sci. 2016;16(1):149-152. 32. Sun E, Liang G, Wang L, et al. Antimicrobial susceptibility of hospital acquired Stenotrophomonas maltophilia isolate biofilms. Braz J Infect Dis. 2016;20(4):365-373. 33. Flamm RK, Castanheira M, Streit JM, Jones RN. Minocycline activity tested against Acinetobacter baumannii, complex, Stenotrophomonas maltophilia, and Burkholderia cepacia species complex isolates from a global surveillance program (2013). Diagn Microbiol Infect Dis. 2016;85:352-355. 34. Wei C, Ni W, Cai X, Zhao J, Cui J. Evaluation of trimethoprim/sulfamethoxazole (SXT), minocycline, tigecycline, moxifloxacin, and ceftazidime alone and in combinations for SXTsusceptible and SXT-resistant Stenotrophomonas maltophilia by in vitro time-kill experiments. PLoS One. 2016;11(3):1-9. 35. Wei C, Ni W, Cai X, Cui J. A monte carlo pharmacokinetic/pharmacodynamics simulation to evaluate the efficacy of minocycline, tigecycline, moxifloxacin, and levofloxacin in the treatment of hospital-acquired pneumonia caused by Stenotrophomonas maltophilia. Infect Dis (Lond). 2015;47(12):846-851. 36. Jia W, Wang J, Xu H, Li G. Resistance of Stenotrophomonas maltophilia to fluoroquinolones: prevalence in a university hospital and possible mechanisms. Int J Environ Res Public Health. 2015;12(5):5177-5195. 37. Cho SY, Lee DG, Choi SM, et al. Stenotrophomonas maltophilia bloodstream infection in patients with hematological malignancies: a retrospective study and in vitro activities of antimicrobial combinations. BMC Infect Dis. 2015;15(69):1-8. 38. Usta E, Eroglu C, Yanik K, et al. Investigation of the presence of class 1, 2, 3 integrons and their relationships with antibiotic resistance in clinical Stenotrophomonas maltophilia isolates. Mikrobiyol Bul. 2015;49(1):35-46. Cosimi 17

39. Sanchez-Borges M, Thong B, Blanca M, et al. Hypersensitivity reactions to non beta-lactam antimicrobial agents, a statement of the WAO special committee on drug allergy. World Allergy Organ J. 2013;6(18):1-23. 40. Nguyen AT, Gentry CA, Furrh RZ. A comparison of adverse drug reactions between high- and standard-dose trimethoprim-sulfamethoxazole in the ambulatory setting. Current Drug Safety. 2013;8:114-119. 41. Fox ER, Beckwith MC. Sulfamethoxazole/trimethopriminjection. ASHP. Updated: April 16, 2014. Accessed: November 8, 2016. http://www.ashp.org/menu/drugshortages/resolved Shortages/bulletin.aspx?id=613. 42. Wang YL, Scipione MR, Dubrovskaya Y, Papadopoulos J. Monotherapy with fluoroquinolone or trimethoprim-sulfamethoxazole for treatment of Stenotrophomonas maltophilia infections. Antimicrob Agents Chemother. 2014;58(1):176-182. 43. Cho SY, Kang C, Kim J, et al. Can levofloxacin be a useful alternative to trimethoprimsulfamethoxazole for treating Stenotrophomonas maltophilia bacteremia? Antimicrob Agents Chemother. 2014;58(1):581-583. 44. Jacobson S, Noa LJ, Wallace MR, Bowman MC. Clinical outcomes using minocycline for Stenotrophomonas maltophilia infections. J Antimicrob Chemother. Doi:10.1093/jac/dkw327. Cosimi 18