TOPICS. Fluoroquinolone Generations: What s Next?

Similar documents
Pathogens and Antibiotic Sensitivities in Post- Phacoemulsification Endophthalmitis, Kaiser Permanente, California,

CME/CE QUIZ CME/CE QUESTIONS. a) 20% b) 22% c) 34% d) 35% b) Susceptible and resistant strains of typical respiratory

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

Topical Antibiotic Update. Brad Sutton, O.D., F.A.A.O. Indiana University School of Optometry Indianapolis Eye Care Center No financial disclosures

Cataracts are the leading cause of visual impairment and

Adoption of intracameral antibiotic prophylaxis of endophthalmitis following cataract surgery: update on the ESCRS Endophthalmitis Study.

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

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

PK/PD to fight resistance

ANTIBIOTICS: TECHNOLOGIES AND GLOBAL MARKETS

Bacterial Resistance of Respiratory Pathogens. John C. Rotschafer, Pharm.D. University of Minnesota

General Approach to Infectious Diseases

Antimicrobial Stewardship in the Hospital Setting

Healthcare Facilities and Healthcare Professionals. Public

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

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

American Association of Feline Practitioners American Animal Hospital Association

Introduction to Chemotherapeutic Agents. Munir Gharaibeh MD, PhD, MHPE School of Medicine, The university of Jordan November 2018

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

EDUCATIONAL COMMENTARY - Methicillin-Resistant Staphylococcus aureus: An Update

COMMITTEE FOR VETERINARY MEDICINAL PRODUCTS

The Battle of Resistance: Treating Infections in the Age of Resistance

Bacterial Resistance. The Battle of the Bugs: Treating Infections in the Age of Resistance. How Resistance Develops. The Age of Modern Medicine

Antimicrobial stewardship in companion animals: Welcome to a whole new era

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

Antimicrobial Stewardship Strategy: Antibiograms

Choosing the Ideal Antibiotic Therapy and the Role of the Newer Fluoroquinolones in Respiratory Tract Infections

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

Optimizing Antimicrobial Stewardship Activities Based on Institutional Resources

Microbiology : antimicrobial drugs. Sheet 11. Ali abualhija

Development and improvement of diagnostics to improve use of antibiotics and alternatives to antibiotics

Abstract... i. Committee Membership... iii. Foreword... vii. 1 Scope Definitions... 1

INOcular. Antiinfectives TOPICS. Keeping Mrsa in check. Francis S. Mah, MD CME

Prophylactic antibiotic timing and dosage. Dr. Sanjeev Singh AIMS, Kochi

Barriers to Intravenous Penicillin Use for Treatment of Nonmeningitis

Objective 1/20/2016. Expanding Antimicrobial Stewardship into the Outpatient Setting. Disclosure Statement of Financial Interest

UPDATE ON ANTIMICROBIAL STEWARDSHIP REGULATIONS AND IMPLEMENTATION OF AN AMS PROGRAM

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

CHSPSC, LLC Antimicrobial Stewardship Education Series

ANTIBIOTIC STEWARDSHIP

Cephalosporins, Quinolones and Co-amoxiclav Prescribing Audit

Visit ABLE on the Web at:

Appropriate antimicrobial therapy in HAP: What does this mean?

No-leaching. No-resistance. No-toxicity. >99.999% Introducing BIOGUARD. Best-in-class dressings for your infection control program

Understanding the Hospital Antibiogram

Methicillin-Resistant Staphylococcus aureus

CHAPTER:1 THE RATIONAL USE OF ANTIBIOTICS. BY Mrs. K.SHAILAJA., M. PHARM., LECTURER DEPT OF PHARMACY PRACTICE, SRM COLLEGE OF PHARMACY

GENERAL NOTES: 2016 site of infection type of organism location of the patient

Principles of Antimicrobial therapy

Recommended for Implementation at Step 7 of the VICH Process on 15 December 2004 by the VICH Steering Committee

For analyst certification and disclosures please see page 7

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

GUIDE TO INFECTION CONTROL IN THE HOSPITAL. Antibiotic Resistance

Public Assessment Report. Scientific discussion. Xiflodrop 5 mg/ml eye drops, solution. Moxifloxacin hydrochloride DK/H/2221/001/DC

International Food Safety Authorities Network (INFOSAN) Antimicrobial Resistance from Food Animals

COMMITTEE FOR MEDICINAL PRODUCTS FOR VETERINARY USE (CVMP) REVISED GUIDELINE ON THE SPC FOR ANTIMICROBIAL PRODUCTS

Antimicrobial Pharmacodynamics

Clinical Guideline. District Infectious Diseases Management. Go to Guideline. District Infectious Diseases Management CG 18_24

All India Ophthalmological Society members survey results: Cataract surgery antibiotic prophylaxis current practice pattern 2017

Baytril 100 (enrofloxacin) Injectable is FDA-approved for BRD control (metaphylaxis) in high-risk cattle.

What Canadian vets need to know and explain about antimicrobial resistance

Study of Bacteriological Profile of Corneal Ulcers in Patients Attending VIMS, Ballari, India

Principles of Antimicrobial Therapy

Treatment of Respiratory Tract Infections Prof. Mohammad Alhumayyd Dr. Aliah Alshanwani

2018 OPTIONS FOR INDIVIDUAL MEASURES: REGISTRY ONLY. MEASURE TYPE: Process

Concise Antibiogram Toolkit Background

مادة االدوية المرحلة الثالثة م. غدير حاتم محمد

Antibacterial Agents & Conditions. Stijn van der Veen

2019 COLLECTION TYPE: MIPS CLINICAL QUALITY MEASURES (CQMS) MEASURE TYPE: Process High Priority

Tel: Fax:

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

ETX0282, a Novel Oral Agent Against Multidrug-Resistant Enterobacteriaceae

INTRACAMERAL PROPHYLAXIS IN CATARACT SURGERY

PDF hosted at the Radboud Repository of the Radboud University Nijmegen

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

Background and Plan of Analysis

Antimicrobial Selection to Combat Resistance

Received: Accepted: Access this article online Website: Quick Response Code:

Considerations in antimicrobial prescribing Perspective: drug resistance

AHFA 2016 Regulatory Summit. Antimicrobial Material Preservatives & Sustainability Considerations

Dr. P. P. Doke. M.D., D.N.B., Ph.D., FIPHA. Professor, Department of Community Medicine, Bharati Vidyapeeth Medical College, Pune

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

Antimicrobial Stewardship 101

Executive Summary: A Point Prevalence Survey of Antimicrobial Use: Benchmarking and Patterns of Use to Support Antimicrobial Stewardship Efforts

Antibiotic Stewardship in the LTC Setting

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

The Basics: Using CLSI Antimicrobial Susceptibility Testing Standards

Antimicrobial Resistance

Antimicrobial Resistance Acquisition of Foreign DNA

The trinity of infection management: United Kingdom coalition statement

TREAT Steward. Antimicrobial Stewardship software with personalized decision support

The Infected Implant in Orthopaedic Reconstruction: An Update on the Clinical and Molecular Approaches to Prevention and Diagnosis

Submission for Reclassification

Commonwealth of Kentucky Antibiotic Stewardship Practice Assessment For Long-Term Care Facilities

2017 Introduction to Infectious Diseases Clinical Seminar Saturday 30th September - Sunday 1st October 2017 Hotel Grand Chancellor Hobart, Tasmania

Lab Exercise: Antibiotics- Evaluation using Kirby Bauer method.

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

Host, Syndrome, Bug, Drug: Introducing 2 Frameworks to Approach Infectious Diseases Cases with an Antimicrobial Stewardship Focus

Does the Dose Matter?

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

Transcription:

A CONTINUING MEDICAL EDUCATION PUBLICATION CME CONTINUING MEDICAL EDUCATION NOVEMBER 2016 ISSUE 63 INOcular TOPICS Antiinfectives Fluoroquinolone Generations: What s Next? Parag A. Majmudar, MD The trend in fluoroquinolone development toward better gram-positive coverage makes later generations particularly valuable for empiric treatment of ocular surface infections. But should the next generation be a new mode of delivery rather than a molecular refinement? A generation is a classification marking an advance in pharmaceutical development within a class of agents. With respect to antibiotics, a modification in chemical structure, a twist in the mechanism of action, a shift or expansion of antimicrobial activity, improved efficacy, lower resistance, better tolerability, altered pharmacokinetics, or some combination of the above, may solve a problem presented by a prior generation of the medication. The class of systemic cephalosporins epitomize the concept of chronologically unfolding generations in antibiotic development, as each new development resulted in a shift in the antimicrobial spectrum toward greater anti-gramnegative and anti-anaerobic activity and improved the range of pharmacokinetic problems associated with the generation before. The result is the diverse catalogue of cephalosporin agents in use today, all using a similar mechanism but with unique antibacterial potencies and properties. The fluoroquinolone class expanded in the opposite microbiologic direction, starting as mostly gram-negative, but progressing to increasing gram-positive coverage. And while the categories or generations are perhaps less clear compared to cephalosporins (eg, it is unclear whether levofloxacin, the levo-isomer of ofloxacin, is a 2nd or 3rd generation fluoroquinolone), the idea is basically the same. 1 The systemic fluoroquinolones originated in the 1960s with naladixic acid, an anti-gram-negative agent excreted in high concentrations in urine and used principally for the treatment of urinary tract infection. Ensuing fluoroquinolone generations employed structural modifications to achieve varying affinities for two target enzymes involved in bacterial DNA replication: DNA gyrase (also called topoisomerase II) and topoisomerase IV. Thus, fluoroquinolone generations evolved from narrow-spectrum antigram-negative activity (1st generation, eg, naladixic acid) to expanded antigram-negative activity including Pseudomonas (2nd generation, eg, ciprofloxacin) to increasing gram-positive fighting activity (3rd generation, eg, levofloxacin; See INSIDE for: Applying MIC in Antibiotic Drug Selection and Resistance Prevention by Joseph M. Blondeau, PhD TARGET AUDIENCE This educational activity is intended for ophthalmologists and ophthalmologists in residency or fellowship training. LEARNING OBJECTIVES Upon completion of this activity, participants will be able to: 1. Factor antibiotic generation into clinical decision-making in the treatment and prevention of ocular infection. 2. Recognize limitations of generational antibiotic categorization. 3. Explain changing trends in antibiotic resistance to common ocular pathogens and the techniques used to establish these. 4. Describe strategies for minimizing antibiotic resistance through changes in clinical practice. EDITORS NISHA ACHARYA, MD, is an associate professor of ophthalmology and epidemiology at the University of California, San Francisco and director of the Uveitis Service at the F.I. Proctor Foundation. NATALIE AFSHARI, MD, FACS, is professor of ophthalmology and chief of cornea and refractive surgery at the Shiley Eye Center, University of California San Diego. MELISSA DALUVOY, MD, is an assistant professor of ophthalmology at the Duke University School of Medicine, and corneal specialist at Duke Eye Center. Topics in Ocular Antiinfectives is jointly sponsored by Candeo Clinical/Science Communications, LLC, and the University of Florida College of Medicine. This publication is administered by an independent editorial board and supported by an unrestricted educational grant from Bausch + Lomb, Inc. Copyright 2016 Candeo Clinical/Science Communications, LLC. All rights reserved. Neither the University of Florida nor Candeo Clinical/Science Communications, LLC, assume any responsibility for injury or damage to persons or property arising from the use of information or ideas contained in this publication. COURSE DIRECTOR MATTHEW J. GRAY, MD University of Florida Gainesville, FL, USA Supported by an unrestricted educational TOPICS IN grant OCULAR from ANTIINFECTIVES Bausch + Lomb, Inc. 1

and 4th generation, eg, gatifloxacin, moxifloxacin, and besifloxacin). Specific core and side chain modifications have also been used to improve tolerability, pharmacokinetic properties, and lower resistance risk. 2,3 Ocular Fluoroquinolones The ocular fluoroquinolones are widely prescribed for their broad antibacterial activity, clinical efficacy, good tolerability, and convenient dosing. Earlier-generation topical ocular fluoroquinolones, including ciprofloxacin and levofloxacin, have excellent broadspectrum coverage and are sometimes preferred to later generation agents for treating ocular infections caused by gram-negative pathogens, including those associated with contact lens wear when Pseudomonas is suspected or documented. 4,5 The trend in fluoroquinolone development toward better gram-positive coverage makes later generations (eg, m o x i fl o x a c i n, g a t i fl o x a c i n, b e s i fl o x a- cin) particularly valuable for empiric treatment of ocular surface infections, a majority of which are due to Staphylococcus, Corynebacterium, and other common gram-positive pathogens. 6 The first fluoroquinolone to contain a chlorine moiety, ie, the first chlorofluoroquinolone, besifloxacin binds to both target enzymes in a more balanced fashion than prior fluoroquinolones, which is important for several reasons. First, it broadens the antimicrobial activity spectrum. Besifloxacin has in vitro activity against gram-negative pathogens, including Pseudomonas, and gram-positive pathogens, including methicillin-resistant Staphylococcus aureus (MRSA). 6,7 This is particularly important, as methicillin resistance has become increasingly prevalent among staphylococcal pathogens in recent decades. 8-12 Second, besifloxacin s dual mode of action reduces risk for the emergence of resistant pathogens since two mutations one in the gene encoding each enzyme would be required to incapacitate the drug. 7 Besifloxacin is also distinctive for being the only antibiotic that is solely available for human topical ocular use; it has not been developed for systemic use in medicine and is not used in livestock or agriculture. 7 Limiting human and Topics in Ocular Antiinfectives, Issue 63 STATEMENT OF NEED Ophthalmologists face numerous challenges in optimizing their competencies and clinical practices in the realm of preventing, diagnosing, and treating ocular infections and their sequelae; these challenges include: The widespread off-label use of topical ophthalmic antibiotics to prevent and treat serious and sight-threatening infections given the reality that the most widely used topical antibiotics in ophthalmology have FDA approvals restricted to bacterial conjunctivitis. The escalating levels of multi-drug resistance in common ocular pathogens. 1 The emergence and increasing prevalence of once-atypical infections that may require diagnostic and treatment techniques relatively unfamiliar to comprehensive ophthalmologists. 2 The introduction of new and potentially more efficacious and/or safe ophthalmic antiinfectives. 3 The introduction of new and potentially more accurate diagnostic techniques for ophthalmic infections. 4 Widespread discussion over the efficacy and safety of novel or alternative delivery techniques and vehicles for prophylactic ophthalmic antibiotics (including but not limited to intracameral injection and topical mucoadhesives). 5,6 Increased understanding of the inflammatory damage caused by ocular infections and the best ways to prevent/ alleviate inflammation without fueling the growth of pathogenic organisms. Given the continually evolving challenges described above, Topics in Ocular Antiinfectives aims to help ophthalmologists update outdated competencies and narrow gaps between actual and optimal clinical practices. As an ongoing resource, this series will support evidence-based and rational antiinfective choices across a range of ophthalmic clinical situations. REFERENCES 1. Asbell PA, Colby KA, Deng S, et al. Ocular TRUST: nationwide antimicrobial susceptibility patterns in ocular isolates. Am J Ophthalmol. 2008 Jun;145(6):951-8. 2. Gower EW, Keay LJ, Oechsler RA, et al. Trends in fungal keratitis in the United States, 2001 to 2007. Ophthalmology. 2010 Dec;117(12):2263-7. 3. Colin J, Hoh HB, Easty DL, et al. Ganciclovir ophthalmic gel (Virgan 0.15%) in the treatment of herpes simplex keratitis. Cornea. 1997;16:393-9. 4. Sambursky R, Tauber S, Schirra F, et al. The RPS adeno detector for diagnosing adenoviral conjunctivitis. Ophthalmology. 2006;113(10):1758-64. 5. Akpek EK, Vittitow J, Verhoeven RS, et al. Ocular surface distribution and pharmacokinetics of a novel ophthalmic 1% azithromycin formulation. J Ocul Pharmacol Ther. 2009;25:433-9. 6. Endophthalmitis Study Group, European Society of Cataract & Refractive Surgeons. Prophylaxis of postoperative endophthalmitis following cataract surgery: results of the ESCRS multicenter study and identification of risk factors. J Cataract Refract Surg. 2007;33(6):978-88. OFF-LABEL USE STATEMENT This work discusses offlabel uses of antiinfective medications. GENERAL INFORMATION This CME activity is sponsored by the University of Florida College of Medicine and is supported by an unrestricted educational grant from Bausch + Lomb, Inc. Directions: Select one answer to each question in the exam (questions 1 10) and in the evaluation (questions 11 16). The University of Florida College of Medicine designates this enduring material for a maximum of 1.0 AMA PRA Category 1 Credit. There is no fee to participate in this activity. In order to receive CME credit, participants should read the report, and then take the posttest. A score of 80% is required to qualify for CME credit. Estimated time to complete the activity is 60 minutes. On completion, tear out or photocopy the answer sheet and send it to: University of Florida CME Office PO Box 100233, Gainesville, FL 32610-0233 phone: 352-733-0064 fax: 352-733-0007 Or you can take the test online at http://cme.ufl.edu/ocular System requirements for this activity are: For PC users: Windows 2000, XP, 2003 Server, or Vista; Internet Explorer 6.0 or newer, or Mozilla Firefox 2.0 or newer ( JavaScript and Java enabled). For Mac users: Mac OS X 10.4 (Tiger ) or newer; Safari 3.0 or newer, Mozilla Firefox 2.0 or newer; ( JavaScript and Java enabled). Internet connection required: Cable modem, DSL, or better. DATE OF ORIGINAL RELEASE November 2016. Approved for a period of 12 months. ACCREDITATION STATEMENT This activity has been planned and implemented in accordance with the accreditation requirements and policies of the Accreditation Council for Continuing Medical Education through the joint providership of the University of Florida College of Medicine and Candeo Clinical/Science Communications, LLC. The University of Florida College of Medicine is accredited by the ACCME to provide continuing medical education for physicians. CREDIT DESIGNATION STATEMENT The University of Florida College of Medicine designates this enduring material for a maximum of 1 AMA PRA Category 1 Credit. Physicians should claim only the credit commensurate with the extent of their participation in the activity. FACULTY AND DISCLOSURE STATEMENTS Nisha Acharya, MD (Faculty Advisor), is an associate professor of ophthalmology and epidemiology at the University of California, San Francisco and director of the Uveitis Service at the F.I. Proctor Foundation. She states that in the past 12 months, she has not had a financial relationship with any commercial organization that produces, markets, re-sells, or distributes healthcare goods or services consumed by or used on patients. Natalie Afshari MD, FACS (Faculty Advisor), is professor of ophthalmology and chief of cornea and refractive surgery at the Shiley Eye Center, University of California San Diego. Dr. Afshari has received grant/research support from the National Institutes of Health, and has served as a consultant for NovaBay Pharmaceuticals, Inc. Melissa Daluvoy, MD (Faculty Advisor), is an assistant professor of ophthalmology at the Duke University School of Medicine, and corneal specialist at Duke Eye Center. She states that in the past 12 months, she has not had a financial relationship with any commercial organization that produces, markets, resells, or distributes healthcare goods or services consumed by or used on patients. Joseph M. Blondeau, PhD, is head of clinical microbiology at Royal University Hospital and the Saskatoon Health Region in Saskatoon, Saskatchewan, Canada. Dr. Blondeau is also associate professor of pathology, adjunct professor of microbiology and immunology, and clinical associate professor of ophthalmology at the University of Saskatchewan. He states that in the past 12 months, he has not had a financial relationship with any commercial organization that produces, markets, resells, or distributes healthcare goods or services consumed by or used on patients relevant to this manuscript. Parag A. Majmudar, MD, is an associate professor of ophthalmology at Rush University Medical Center, and in private practice at Chicago Cornea Consultants. Dr. Majmudar is a consultant for Alcon, Valeant Pharmaceuticals, and Shire. DISCLAIMER Participants have an implied responsibility to use the newly acquired information to enhance patient outcomes and professional development. The information presented in this activity is not meant to serve as a guideline for patient care. Procedures, medications, and other courses of diagnosis and treatment discussed or suggested in this activity should not be used by clinicians without evaluation of their patients conditions and possible contraindications or dangers in use, applicable manufacturer s product information, and comparison with recommendations of other authorities. COMMERCIAL SUPPORTERS This activity is supported by an unrestricted educational grant from Bausch + Lomb, Inc. 2 TOPICS IN OCULAR ANTIINFECTIVES

environmental exposure to besifloxacin is a strategy for reducing selective pressure for the development of resistance and prolonging its usefulness as an effective ocular antibiotic, a strategy that may in fact be working. According to recent large microbiologic surveys of topical ocular pathogens, besifloxacin resistance remains low. 13 Ongoing surveillance will shed light on whether this is an enduring phenomenon and, if so, whether besifloxacin s unique chemical structure and environmental containment are contributing to its longevity. The Real World There is some merit to considering antibiotic generation in therapeutic decision-making. Opting for a 3rd generation fluoroquinolone is reasonable when a gram-negative pathogen is suspected, and opting for a 4th generation fluoroquinolone is reasonable when Staphylococcus is more likely. An argument could be made, however, for exclusively using latest-generation agents in clinical practice, as they represent the most advanced iteration of a drug class and are often associated with minimal resistance. Furthermore, later-generation antibiotics are better supported by drug manufacturers, which means that samples are in greater supply. Still, a generation is something of an artificial designation. In real world ophthalmic practice, selecting antibiotics by generation may be beside the point. Other considerations such as pharmacodynamic or practical ones may be of greater importance. For example, later-generation antibiotics generally demonstrate greater in vitro potency against resistant pathogens; however, this advantage may be clinically unimportant given the fact that minimal inhibitory concentrations (MICs) reflect organism susceptibility to drug concentrations in the bloodstream, not on the ocular surface. 14 In general, the concern around antibiotic resistance even resistance that is widespread and increasing, as with MRSA poses less of a threat to the eye, since antibiotic concentrations achievable on the eye s surface are orders of magnitude higher than those achievable in the blood or other anatomical sites and likely well above the MIC even for resistant pathogens. In addition, earlier-generation antibiotics, often dispensed in generic form, may be sufficient for what has become a common, albeit controversial, use for topical ocular antibiotics: prophylaxis around intravitreal injection and intraocular surgery. While the practice of applying topical antibiotics prior to injection or surgery has been shown to reduce bacterial counts on the surface, it has never been shown to reduce the incidence of endophthalmitis over povidone-iodine antisepsis alone. 15,16 The vast majority perhaps upwards of 90% of topical ocular antibiotics prescribed today are dispensed as generic formulations, either because they were prescribed as generic or because a substitution was made by the pharmacist for the purpose of controlling costs. This is a dramatic increase in generic usage compared to 5 to 10 years ago. But widespread use of older and/or generic antibiotics in ophthalmology is not necessarily cause for alarm, as noted above: the ability to achieve high concentrations of antibiotic on the ocular surface enhances efficacy even against pathogens with high MICs; and topically applied antibiotics for prophylactic use may not be necessary in the first place. Future Generations It is unclear exactly what role topical antibiotics will play in ophthalmology in the coming years. Research may help clarify whether or not topical antibiotics are beneficial as infection prophylaxis. Topical ocular antibiotics could lose ground for commercial reasons: for example, the development of new antibiotic molecules is impeded by a lack of financial incentive within the pharmaceutical industry to develop drugs indicated for intermittent, shortterm use and whose efficacy is always at least theoretically threatened by the emergence of resistance. Furthermore, there is a growing trend in favor of new modes of delivery, such as intracameral antibiotic injection at intraocular surgery as a means for preventing endophthalmitis. 17 Th e CORE CONCEPTS Antibiotic generations are evolutions in drug design aimed at filling an unmet need within class of agents. An effective medication advance resolves pertinent problems doctors and patients face. Ocular fluoroquinolones have evolved toward greater gram-positive activity, broader overall coverage, lower susceptibility to resistance pressure, and more effective pharmacokinetic profiles. European Society of Cataract and Refractive Surgery s landmark study of intracameral prophylaxis in 2007 showed a 5-fold decrease in endophthalmitis after cataract surgery with the use of intracameral cefuroxime. 17 Th e results were subsequently corroborated in a number of other large, prospective studies by various groups from Europe and Asia. 18,19 The first comparative US study on intracameral prophylaxis was published in 2013, showing a 10-fold decline in endophthalmitis rates with the routine use of intracameral injection of cefuroxime, moxifloxacin, or vancomycin. 20 Intraoperative antibiotic prophylaxis is also attractive since it eliminates compliance issues and reduces patients post-operative drop burden. With additional prospective studies in the US unlikely to be performed (due to high costs associated with a study of sufficient size to achieve statistical significance of such a rare occurrence), an increasing proportion of surgeons are satisfied with the evidence that exists in support of intracameral prophylaxis and are actively adopting the practice of their European colleagues. But, in the absence of an antibiotic approved for intracameral use, practical and regulatory issues remain. An ideal next-generation ophthalmic antibiotic would not necessarily be a new molecule: it could be a new system that overcomes current practical barriers to more TOPICS IN OCULAR ANTIINFECTIVES 3

widespread adoption of intracameral antibiotic prophylaxis. One potential solution might be commercial development of a cataract kit containing an approved unit-dose, preservative-free injectable antibiotic, such as moxifloxacin or cefuroxime; and injectible antiinflammatory agents, packaged and marketed specifically for ophthalmic surgery prophylaxis. Such a product would free hospital administrators from tedious and costly workarounds currently required to obtain intracameral antibiotics, would be simpler and more expedient for surgeons, and would quite possibly be safer for surgical patients. Conclusion Generations are ways of building on past successes in antibiotic development. When we understand the problem each new generation is trying to surmount, we make more informed clinical decisions. Parag A. Majmudar, MD, is an associate professor of ophthalmology at Rush University Medical Center, and in private practice at Chicago Cornea Consultants. Dr. Majmudar is a consultant for Alcon, Valeant Pharmaceuticals, and Shire. Medical writer Noelle Lake, MD, assisted in the preparation of this manuscript. REFERENCES 1. Young, M. ASCRS Eye World. Fluoroquinolone Generations. 2003. www.eyeworld.org/ article.php? sid=811&strict=0&morphologic =0&query=. Accessed July 25, 2016. 2. Scoper SV. Review of third-and fourthgeneration fluoroquinolones in ophthalmology: in-vitro and in-vivo efficacy. A d v Th e r. 2008;25:979-4. 3. Ball P. Quinolone generations: natural history or natural selection? J Antimicrob Chemother. 2000;46:17-24. 4. Ciprofloxacin Hydrochloride Ophthalmic Solution prescribing information. 2012. Alcon, Princeton, NJ. 5. Levofloxacin Ophthalmic Solution 0.5% prescribing information. 2007. Apotex, Inc. Toronto, CN. 6. Besivance (besifloxacin ophthalmic suspension) 0.6% prescribing information. 2009. Bausch and Lomb. Tampa, FL. 7. Mah FS, Sanfi lippo CM. Besifloxacin: efficacy and safety in treatment and prevention of ocular bacterial Infections. Ophthalmol Ther. 2016;5:1-20. 8. Solomon R, Donnenfeld ED, Holland EJ, et al. Microbial keratitis trends following refractive surgery: results of the ASCRS infectious keratitis survey and comparisons with prior ASCRS surveys of infectious keratitis following keratorefractive procedures. J Cataract Refract Surg. 2011;37:1343-50. 9. Cavuoto K, Zutshi D, Karp CL, et al. Update on bacterial conjunctivitis in South Florida. Ophthalmology. 2008;115:51-6. 10. Major JC Jr, Engelbert M, Flynn HW Jr, et al. Staphylococcus aureus endophthalmitis: antibiotic susceptibilities, methicillin resistance, and clinical outcomes. Am J Ophthalmol. 2010;149(2):278-83. 11. Asbell PA, Sahm DF, Shaw M, et al. Increasing prevalence of methicillin resistance in serious ocular infections caused by Staphylococcus aureus in the United States: 2000 to 2005. J Cataract Refract Surg. 2008;34(5):814-18. 12. Haas W, Pillar CM, Torres M, et al. Monitoring antibiotic resistance in ocular microorganisms: results from the Antibiotic Resistance in Ocular Microorganisms (ARMOR) 2009 Surveillance Study. Am J Ophthalmol. 2011;152:567-74. 13. Asbell PA, Sanfilippo CM, Pillar CM, DeCory HH, Sahm DF, Morris TW. Antibiotic resistance among ocular pathogens in the United States: five-year results from the ARMOR surveillance study. JAMA Ophthalmol. 2015;133:1445-54. 14. Kowalski RP. Is antibiotic resistance a problem in the treatment of ophthalmic infections? Expert Review of Ophthalmology. 2013;8:119-26. 15. Schimel AM, Flynn HW. Infection: the evolving role of antibiotics. Rev Ophthalmology. 2012(Aug):1-6. 16. Speaker MG, Menikoff JA: Prophylaxis of endophthalmitis with topical povidone-iodine. Ophthalmology. 1991;98:1769-75. 17. Endophthalmitis Study Group, European Society of Cataract & Refractive Surgeons. Prophylaxis of postoperative endophthalmitis following cataract surgery: results of the ESCRS multicenter study and identification of risk factors. J Cataract Refract Surg. 2007;33:978-88. 18. Garcia-Saenz MC, Arias-Puente A, Rodriguez-Caravaca G, et al. Effectiveness of intracameral cefuroxime in preventing endophthalmitis after cataract surgery Ten-year comparative study. J Cataract Refract Surg. 2010;36:203-7. 19. Tan CS, Wong HK, Yang FP. Epidemiology of postoperative endophthalmitis in an Asian population: 11-year incidence and effect of intracameral antibiotic agents. J Cataract Refract Surg. 2012;38:425-30. 20. Shorstein NH, Winthrop KL, Herrinton LJ. Decreased postoperative endophthalmitis rate after institution of intracameral antibiotics in a Northern California eye department. J Cataract Refract Surg. 2013;39:8-14. BLONDEAU REFERENCES continued from page 7 5. Johnson CC. In vitro testing: correlations of bacterial susceptibility, body fluid levels and effectiveness of antibacterial therapy. In: Antibiotics in Laboratory Medicine. Lorian V (Ed.). Williams and Wilkins, MA, USA. 813-34 (1996). 6. Hesje CK, Tillotson GS, Blondeau JM. MICs, MPCs and PK/PDs: a match (sometimes) made in hosts. Expert Rev Resp Med. 2007;1(1):7-16. 7. Dong Y, Zhao X, Domagala J, Drlica K. Effect of fluoroquinolone concentration on selection of resistant mutants of Mycobacterium bovis BCG and Staphylococcus aureus. Antimicrob Agents Chemother. 1999;43:1756-8. 8. Hedlin P, Blondeau JM. Comparative minimal inhibitory and mutant prevention drug concentrations of four fluoroquinolones against ocular isolates of haemophilus influenza. Eye Contact Lens. 2007;33(3):161-4. 9. Blondeau JM, Borsos S, Hesje CK. Antimicrobial efficacy of gatifloxacin and moxifloxacin with and without benzalkonium chloride compared with ciprofloxacin and levofloxacin against methicillin-resistant staphylococcus aureus. J Chemother. 2007;19(2):146-51. 10. Hesje CK, Borsos SD, Blondeau JM. Benzalkonium chloride enhances antibacterial activity of gatifloxacin and reduces its propensity to select for fluoroquinolone-resistant strains. J Ocular Pharmacol Therapeut. 2009;25(4):329-33. 4 TOPICS IN OCULAR ANTIINFECTIVES

Applying MIC in Antibiotic Drug Selection and Resistance Prevention Joseph M. Blondeau, PhD A greater understanding of minimum inhibitory concentration and other microbiological measurements can help ophthalmologists select appropriate antibiotic treatment and prevent antibiotic resistance to ocular pathogens. Antibiotic resistance to bacterial pathogens is an escalating public health concern across a number of medical specialties, ophthalmology included. The evolution of pathologic organisms from susceptible to increasingly less responsive to antibiotic treatment can complicate ophthalmologists management strategies for ocular infections and ultimately threaten patients vision. This phenomenon has resulted from the misuse and perhaps overuse of antibiotics to treat staphylococci and streptococci (among other pathogens) responsible for the etiology of ocular infections such as keratitis, conjunctivitis, blepharitis and endophthalmitis. Resistance is likely exacerbated by increased use of the same antibiotics for non-ocular conditions such as respiratory tract infections, which share many of the same pathogens responsible for ocular infection, including Haemophilus influenza, S. pneumoniae, and Pseudomonas aeruginosa. Resistance Surveillance To facilitate understanding of ocular antibiotic resistance patterns in the US and generate geographic-specific information on emerging resistance, two surveillance programs have been implemented: Ocular Tracking Resistance in US Today (TRUST), 1 and more recently the Antibiotic Resistance Monitoring in Ocular Microorganisms (ARMOR) study. 2 Over a period of 20 years, these studies have been gathering data on resistance of common pathogens to ocular antibiotics based on in vitro susceptibility testing of ocular isolates sent annually from participating clinics throughout all 50 states (Figure 1). FIGURE 1 Kirby-Bauer disk diffusion susceptibility test. The diameter of the grey zone of inhibition around a disk is measured to determine bacterial susceptibility or resistance to an antibiotic. Disks around which there is no zone of inhibition indicate that bacteria are resistant to the antibiotic. (Image courtesy of Dr. Blondeau.) The early TRUST data demonstrated that, after more than a decade of systemic use, fluoroquinolones remained active in vitro in approximately 80% of ocular methicillin-sensitive S. aureus (MSSA) isolates, 90% to 100% of S. pneumoniae isolates (100% susceptibility with gatifloxacin, levofloxacin, and moxifloxacin, and approximately 90% susceptibility with ciprofloxacin), and 100% of H. influenza isolates. In contrast, fluoroquinolones were active in only about 15% of methicillin-resistant S. aureus (MRSA) isolates, suggesting the need to consider alternative antibiotics for this pathogen (Figure 2). 1 The much later ARMOR data showed in vitro methicillin resistance in 42% of S. aureus isolates and about 50% of coagulase-negative staphylococci (CNS) isolates and that these also had a high probability of concurrent resistance to the fluoroquinolones, aminoglycosides, CORE CONCEPTS The rise in antibiotic resistance of ocular pathogens to commonly used drugs such as the fluoroquinolones requires the implementation of new strategies to effectively manage patients presenting with infection. Large-scale, US-wide surveillance studies such as TRUST and ARMOR can provide treating ophthalmologists with evolving information on patterns of antibiotic resistance to pathogens typically observed in the clinic and how these differ among geographic regions. An increased understanding of MIC and MPC to predict clinical efficacy of therapeutic drug classes for common ocular pathogens is valuable for making antibiotic choices. Strategies to prevent antibiotic resistance include use of local laboratories for in vitro testing of infective isolates beyond refractory cases, strict adherence to drug manufacturers prescribing guidelines, and patient education on treatment compliance. or macrolides (87% in MRSA isolates and 77% in methicillin-resistant CNS isolates). While all staphylococcal isolates remained susceptible to vancomycin, resistance among S. pneumoniae isolates was highest for azithromycin (34%), and resistance among P. aeruginosa and H. influenzae was low against the various antibiotics tested. 2 TOPICS IN OCULAR ANTIINFECTIVES 5

With respect to geographic trends, ARMOR revealed small differences across the US in susceptibility among S. aureus, S. pneumoniae, and P. aeruginosa isolates, with gram-positive isolates from the West having the lowest rates of resistance, and S. aureus isolates in the South having the highest resistance. 2 These data demonstrate that antibacterial resistance is becoming prevalent among common bacterial causes of ocular infections such as CNS and S. aureus. The 3rd-generation fluoroquinolone antibiotics (ciprofloxacin, ofloxacin, and levofloxacin), which have long been the mainstay for treating ocular infections due to their broad-spectrum antibiotic effect and high in vitro potency, are becoming increasingly less active against gram-positive and gram-negative organisms. 1,3 This is particularly true in MRSA infections which, while a less prevalent cause of ocular infection, are showing an increasingly more severe clinical course than the MSSA strains. The newer 4th-generation fluoroquinolones (moxifloxacin and gatifloxacin) demonstrate broad-spectrum antibiotic effect against both gram-positive and gram-negative organisms; however, ongoing surveillance is also showing some MRSA resistance to these agents, and preservation of their efficacy in targeting ocular pathogens is of paramount importance. Measuring Antibiotic Potential The internationally accepted measurement of the potential efficacy of an antibiotic is minimum inhibitory concentration (MIC). Defined as the lowest concentration of an antimicrobial drug that is required to inhibit visible growth of 105 colony-forming units (CFU) following in vitro incubation for 18 to 24 hours, MIC is used to confirm antibiotic resistance of existing antimicrobial drugs and establish effective concentrations of new drugs. 4 By using either the broth microdilution test, agar dilution test, or Epsilometer test, laboratories compare the new MIC values with previously established drug breakpoint concentrations, known achievable and FIGURE 2 Chromogenic agar plate selective for MRSA, indicated by the denim blue color. (Image courtesy of Dr. Blondeau.) sustained drug concentrations within the host, pharmacokinetic and pharmacodynamic data, and drug toxicity data to determine pathogenic bacteria susceptibility or resistance to the antibiotic under investigation and thus potential for clinical success or failure (Figure 3). 5 In vitro testing does not always translate absolutely to clinical outcomes, as several other factors influence a patient s response to infection especially his or her natural capacity for defense. In addition, MIC evaluations may be more valuable for patients with self-limiting, mild to moderate infection. The most significant limitation of in vitro MIC testing on its predictive value may be that it uses a static density of 105 CFU per ml; this can result in overestimation of the drug concentration required to exert a satisfactory biological response in instances where organism density of an infection is lower than 105 and underestimation of the drug concentration required where organism density is higher than 105. 6 Nevertheless, MIC FIGURE 3 Epsilometer test to determine MIC, which is read at the point where the organism growth intersects the test strip. (Image courtesy of Dr. Blondeau.) remains the gold standard for in vitro susceptibility testing of pathogens to antibiotics and provides a valuable guide for physicians in managing the majority of patients. The mutant prevention concentration (MPC) test addresses some of the limitations associated with MIC testing. 6 This novel technique not only measures a much higher density of organisms than MIC and determines the drug concentration required to inhibit the least susceptible cell present within the population, but it also measures the frequency with which a microbe mutates and develops resistance. 7 Th e drug concentration required to block the mutant cells (ie, the MPC) is typically higher than the MIC drug concentration with the mutant selection window, a so-called danger zone for the selective amplification of resistant subpopulations falling between these. 6 Although more demanding and less adaptable to routine clinical settings, MPC is a valuable tool when used in conjunction with MIC. 8 PK and PD Data The application of pharmacokinetics (PK) and pharmacodynamics (PD) to MIC and MPC outcomes is important in characterizing the antibacterial activity of compounds. PK data define the fate of a drug in the body (eg, its absorption, transformation, distribution, and elimination), while PD data define the effect of a drug on the body and infecting organism (eg, its mechanism of action and efficacy). Drug curves, for example, provide PK and PD data on the peak or maximum drug concentration (C max ), the trough concentration, area under the curve (AUC), and the drug s half-life (which determines dosing frequency). 6 Antimicrobial compounds may be classified as concentrationdependent agents, for which C max :MIC and AUC:MIC ratios are important predictors of outcome with antimicrobial treatment; or time-dependent agents, for which the time that the drug concentration remains in excess of the MIC is important to determining its 6 TOPICS IN OCULAR ANTIINFECTIVES

antimicrobial activity. ingly available through the TRUST and ARMOR surveillance studies) to help tailor management strategies, be aware of specific drug toxicity data that may influence choice of drug for patients with particular medical histories, and, finally, have an understanding of drug constituents beyond the active ingredient. For example, the preservative benzalkonium chloride (BAK) may confer intrinsic bacteriocidal activity against predominantly gram-positive species, including MRSA. 9,10 In vitro studies have shown that, when added to gatifloxacin and moxifloxacin, BAK substantially lowers their MICs and MPCs against MRSA. Preventing Antibiotic Resistance The emergence of antimicrobial resistance of ocular pathogens suggest that new strategies must be employed to inappropriate use of antibiotics. One strategy is for ophthalmologists to utilize their local microbiology lab for testing organism susceptibility and resistance in patient isolates to therapeutic agents. Currently, this is not an uncommon practice for refractory cases; however, it may be valuable to expand this to manage routine cases. In addition, it is important for ophthalmologists to let the testing laboratory know of the types of microbes that are common in ocular samples, as some ocular pathogens, such as S. epidermis or CNS, are commonly considered contaminants in other systemic samples and thus often ignored. To this end, establishing dialogue to modify existing culturing procedures at the laboratory so they are relevant to the identification of ocular pathogens is an important aspect in utilizing in vitro testing laboratories. Of course, it is also important that treating ophthalmologists prescribe the antibiotic of choice in accordance with the manufacturer s instructions ie, at the correct dose and for the correct length of time and that patients are informed of the consequences of noncompliance with these instructions (as is often the case when the condition Choosing an Antibiotic MIC, MPC, and PK/PD results from in vitro testing of antimicrobial agents are fundamental in supporting ophthalmologists to design appropriate treatment strategies for patients to treat ocular infections. Ophthalmologists should consider whether the chosen antibiotic demonstrates sufficient spectrum of activity to cover all potential infection-causing pathogens and whether the administered ocular dose is of adequate concentration to exceed the anticipated MIC/MPC of the drug for those specific organisms. Keep in mind that the MIC and MPC used to measure antibiotic susceptibility to resistance apply to systemic use of antibiotics, which may not always be appropriate for topical (eg, ocular) antibiotic use, where higher concentrations are more typically administered. Resistance to an antibiotic is based on a drug concentration breakpoint, whereby an organism with an MIC at or above that breakpoint is considered resistant. Breakpoints are based on several parameters, including the systemic drug concentration achievable with approved dosing. Drug concentration breakpoints are not calculated for topical antimicrobials, but bacterial strains can still be classified as low-level or high-level in terms of their resistance for the purposes of choosing topical antimicrobials. Low-level resistance means the MIC is close to the (systemic) drug concentration breakpoint, whereas high-level resistance means the MIC is many times higher than the (systemic) drug concentration breakpoint. Organisms of high-level resistance are unlikely to respond to topical antimicrobials if the MIC is very high. In vitro susceptibility testing provides clinical useful information on susceptibility/resistance and drug use, including for topically applied drugs. It is also valuable for ophthalmologists to become familiar with antibiotic resistance trends in their geographical region (such as those becoming increasstarts to resolve). An increasing number of clinical studies are addressing the question of whether shorter dosing periods may be as effective as the currently longer courses of therapy in systemic infectious disease areas, such as for urinary tract infections, and such information would be valuable in the field of ophthalmology. Conclusion The rise of antibiotic resistance to common ocular infections threatens to diminish the clinical utility of important classes of drugs and ultimately impact patient vision. By increasing awareness of evolving resistance patterns and implementing in-clinic strategies to better target pathogens, ophthalmologists may help preserve drug utility for the benefit of present and future patients. Joseph M. Blondeau, PhD, is head of clinical microbiology at Royal University Hospital and the Saskatoon Health Region in Saskatoon, Saskatchewan, Canada. Dr. Blondeau is also associate professor of pathology, adjunct professor of microbiology and immunology, and clinical associate professor of ophthalmology at the University of Saskatchewan. He states that in the past 12 months, he has not had a financial relationship with any commercial organization that produces, markets, resells, or distributes healthcare goods or services consumed by or used on patients relevant to this manuscript. Medical writer, Caroline M. Markey, PhD, of Markey Medical Consulting Pty Ltd, assisted in the preparation of this manuscript. REFERENCES 1. Asbell PA, Colby KA, Deng S, McDonnell P, Meisler DM, Raizman MB, Sheppard JD, Sahm DF. Ocular TRUST: Nationwide antimicrobial susceptibility patterns in ocular isolates. Am J Ophthalmol. 2008;145:951-8. 2. Asbell PA, Sanfilippo CM, Pillar CM, DeCory HH, Sahm DF, Morris TW. Antibiotic resistance among ocular pathogens in the United States five-year results from the antibiotic resistance monitoring in ocular microorganisms (ARMOR) surveillance study. JAMA Ophthalmol. 2015; 133(12):1445-54. 3. Aldred KJ, Kerns RJ, Osheroff N. Mechanisms of quinolone action and resistance. Biochem. 2014;53:1565-74. 4. Andrews JM. Determination of minimum inhibitory concentrations. J Antimicrob Chemo. 2001;48:Suppl.S1,5-16. BLONDEAU REFERENCES continue on page 4 TOPICS IN OCULAR ANTIINFECTIVES 7

EXAMINATION QUESTIONS TOPICS IN OCULAR ANTIINFECTIVES, ISSUE 63 This CME program is sponsored by the University of Florida College of Medicine and supported by an unrestricted educational grant from Bausch + Lomb, Inc. DIRECTIONS: Select the one best answer to each question in the Exam (Questions 1 10) and in the Evaluation (Questions 11 16) below by circling one letter for each answer. Participants must score at least 80% on the questions and complete the entire Evaluation section on the form below. The University of Florida College of Medicine designates this enduring material for a maximum of 1.0 AMA PRA Category 1 Credit. There is no fee to participate in this activity. You can take the test online at http://cme.ufl.edu/ocular. 1. The mutant selection window: A. Reflects the window of drug concentration required to inhibit visible growth of 105 CFU following in vitro incubation B. Is the danger zone for the selective amplification of resistant subpopulations C. Is the most important predictor of outcome for antimicrobial treatment D. Is typically lower than the MIC drug concentration 2. First-generation quinolone naladixic acid was principally used to treat: A. UTI B. Otitis media C. Colitis D. Soft tissue infection 3. Which statement regarding MIC is FALSE? A. MIC is defined as the lowest concentration of an antimicrobial drug that is required to inhibit visible growth of 105 CFU following in vitro incubation for 18 to 24 hours. B. MIC is a valuable tool for determining effective concentrations of drugs for systemic infections C. MIC measures the frequency of a microbe to mutate and develop resistance D. The ratio of drug area under the curve (AUC) to MIC provides important information to predict outcomes with antimicrobial treatment 4. Which of the following advancements are present in later-generation ophthalmic fluoroquinolones? A. Greater anti-gram-negative activity B. Higher risk for resistance selection C. More balanced MOA D. None of the above 5. Which of the following has NOT been proven to reduce the incidence of postoperative endophthamitis following intraocular surgery? A. Povidone-iodine prep B. Topical ocular moxifloxacin prophylaxis C. Intracameral cefuroxime prophylaxis D. All of the above are proven to prevent endophthalmitis 6. Which statement is TRUE regarding the addition of excipients to antibiotic formulations? A. Higher MIC and MPC values are observed when benzalkonium chloride is added to gatifloxacin and moxifloxacin B. Lower MIC and MPC values against MRSA are observed when BAK is added to gatifloxacin and moxifloxacin C. Higher MIC and MPC values are observed when polyethylene glycol is added to moxifloxacin D. Mannitol increases the in vitro activity of gatifloxacin 7. Which statement is TRUE regarding the key findings of the surveillance studies TRUST and ARMOR? A. Fluoroquinolones remain active in the majority of MRSA isolates B. In vitro S. aureus and CNS isolates have a high probability of resistance to the fluoroquinolones aminoglycosides and or macrolides C. TRUST data show that fluoroquinolone activity has been significantly diminished in the majority of MSSA, S. pneumonia, and H. influenza isolates D. Vancomycin remains active in staphylococcal isolates, but the fourthgeneration fluoroquinolones (moxifloxacin and gatifloxacin) demonstrate decreased spectrum antibiotic effect against both the grampositive and gram-negative organisms 8. Which of the following may be modified in a new generation of antibiotic? A. Chemical structure B. Antimicrobial activity C. Resistance profile D. Any of the above may be modified 9. Fluoroquinolones target which of the following bacterial enzymes? A. DNA gyrase B. Topoisomerase IV C. Beta-lactamase D. A and B 10. Recommended strategies for minimizing antibiotic resistance: A. Include in vitro testing of refractory and routine cases B. Strict adherence to drug manufacturers prescribing information, and patient education on the importance of treatment compliance C. Increasing the awareness of evolving resistance patterns D. All of the above EXAMINATION ANSWER SHEET TOPICS IN OCULAR ANTIINFECTIVES, ISSUE 63 This CME activity is jointly sponsored by the University of Florida and Candeo Clinical/Science Communications, LLC, and supported by an unrestricted educational grant from Bausch + Lomb, Inc. Mail to: University of Florida CME Office, PO Box 100233, Gainesville, FL 32610-0233. DIRECTIONS: Select the one best answer for each question in the exam above (Questions 1 10). Participants must score at least 80% on the questions and complete the entire Evaluation (Questions 11 16) to receive CME credit. CME exam expires October 31, 2017. ANSWERS: 1. A B C D 6. A B C D EVALUATION: 1=Poor 2=Fair 3=Satisfactory 4=Good 5=Outstanding 11. Extent to which the activity met the identified: Objective 1: 1 2 3 4 5 Objective 2: 1 2 3 4 5 Objective 3: 1 2 3 4 5 Objective 4: 1 2 3 4 5 12. Rate the overall effectiveness of how the activity: Related to my practice: 1 2 3 4 5 Will influence how I practice: 1 2 3 4 5 Will help me improve patient care: 1 2 3 4 5 Stimulated my intellectual curiosity: 1 2 3 4 5 Overall quality of material: 1 2 3 4 5 Overall met my expectations: 1 2 3 4 5 Avoided commercial bias/influence: 1 2 3 4 5 If you wish to receive credit for this activity, please fill in the following information. Retain a copy for your records. PLEASE PRINT CLEARLY FIRST NAME LAST NAME DEGREE ORGANIZATION/INSTITUTE ADDRESS LINE 1 ADDRESS LINE 2 2. A B C D 7. A B C D 13. Will the information presented cause you to make any changes in your practice? Yes No CITY STATE ZIP 3. A B C D 8. A B C D 14. If yes, please describe: 4. A B C D 9. A B C D PHONE FAX 15. How committed are you to making these changes? 5. A B C D 10. A B C D 1 2 3 4 5 16. Are future activities on this topic important to you? E-MAIL ADDRESS 8 TOPICS IN OCULAR ANTIINFECTIVES Yes No