Você está na página 1de 69

Volume 9 • Number 4 • July 2010

NephSAP
®

Nephrology Self-Assessment Program

Clinical Pharmacology for


the Nephrologist
Guest Co-Editors:
Suzanne K. Swan, MD,
Ali Olyaei, PharmD, and
Domenic A. Sica, MD
■ Editor-in-Chief: Stanley Goldfarb, MD
■ Deputy Editor: Jeffrey S. Berns, MD
NephSAP
®

EDITOR-IN-CHIEF
Stanley Goldfarb, MD
University of Pennsylvania Medical School
Preface
Philadelphia, PA NephSAP® is one of the three major publications of the American Society of Nephrology
(ASN). Its primary goals are self-assessment, education, and the provision of Continuing
DEPUTY EDITOR Medical Education (CME) credits and Maintenance of Certification (MOC) credits for
Jeffrey S. Berns, MD individuals certified by the American Board of Internal Medicine. Members of the ASN
University of Pennsylvania Medical School
Philadelphia, PA
automatically receive NephSAP with their monthly issue of The Journal of the American
Society of Nephrology (JASN).
MANAGING EDITOR
EDUCATION: Medical and Nephrologic information continually accrues at a rapid pace.
Gisela Deuter, BSN, MSA
Washington, DC Bombarded from all sides with demands on their time, busy practitioners, academicians,
and trainees at all levels are increasingly challenged to review and understand all this new
ASSOCIATE EDITORS material.
Rajiv Agarwal, MD Each bimonthly issue of NephSAP is dedicated to a specific theme, i.e., to a specific
Indiana University School of Medicine area of clinical nephrology, hypertension, dialysis, and transplantation, and consists of an
Indianapolis, IN
Editorial, a Syllabus, a Commentary on the Syllabus, and self-assessment questions. Over
David J. Cohen, MD
Columbia University the course of 24 months, all clinically relevant and key elements of nephrology will be
New York, NY reviewed and updated. The authors of each issue digest, assimilate, and interpret key
Michael Emmett, MD publications from the previous issues of other years and integrate this new material with the
Baylor University body of existing information.
Dallas, TX
Steven Fishbane, MD SELF-ASSESSMENT: Twenty-five single-best-answer questions will follow the 50 to 75 pages
Stony Brook School of Medicine of Syllabus text. The examination is available online with immediate feedback. Those answer-
Minneola, NY ing ⬎75% correctly will receive CME credit, and receive the answers to all the questions along
Richard J. Glassock, MD with brief discussions and an updated bibliography. To help answer the questions, readers may
Professor Emeritus, The David Geffen School
of Medicine at the University of California
go to the ASN web site, where relevant material from UpToDate in nephrology will be posted.
Los Angeles, CA Thus, members will find a new area reviewed every 2 months, and they will be able to test their
Kevin J. Martin, MBBCh understanding with our quiz. This format will help readers stay abreast of developing areas of
St. Louis University School of Medicine clinical nephrology, hypertension, dialysis, and transplantation, and the review and update will
St. Louis, MO support those taking certification and recertification examinations.
Rajnish Mehrotra, MD
Harbor UCLA Research and Education Institute CONTINUING MEDICAL EDUCATION: Most state and local medical agencies as well as
Torrance, CA hospitals are demanding documentation of requisite CME credits for licensure and for staff
Patrick T. Murray, MD appointments. A maximum of 36 credits annually can be obtained by successfully completing
University College Dublin the NephSAP examination. In addition, individuals certified by the American Board of Internal
Dublin, Ireland
Medicine may obtain credits towards Maintenance of Certification (MOC) by successfully
Patrick H. Nachman, MD
University of North Carolina completing the self-assessment portion of NephSAP.
Chapel Hill, NC
BOARD CERTIFICATION AND INSERVICE EXAMINATION PREPARATION: Each issue
Paul M. Palevsky, MD
University of Pittsburgh School of Medicine
will also contain 5 questions and answers examining core topics in the particular discipline
Pittsburgh, PA reviewed in the Syllabus. These questions are designed to provide trainees with challenging
Richard H. Sterns, MD questions to test their knowledge of key areas of nephrology.
University of Rochester School of Medicine
and Dentistry ⬁ This paper meets the requirements of ANSI/NISO Z39.48-1921 (Permanence of Paper),
Rochester, NY effective with July 2002, Vol. 1, No. 1.
Stephen C. Textor, MD
Mayo Clinic
Rochester, MN GUEST CO-EDITORS Suzanne K. Swan, MD
Raymond R. Townsend, MD Ali Olyaei, PharmD Hennepin County Medical Center
University of Pennsylvania Medical School Oregon Health and Science University Minneapolis, MN
Philadelphia, PA Portland, OR
FOUNDING EDITORS
John P. Vella, MD Domenic A. Sica, MD
Maine Medical Center Richard J. Glassock, MD, MACP
Virginia Commonwealth University Editor-in-Chief Emeritus
Portland, ME Richmond, VA
Robert G. Narins, MD, MACP

NephSAP®
©2010 by The American Society of Nephrology
NephSAP
®
Volume 9, Number 4, July 2010

Clinical Pharmacology for the Nephrologist

Editorial 213
Pharmacology and Kidney Disease: Lessons Learned from the
Gadolinium–Nephrogenic Systemic Fibrosis Story
—Mark A. Perazella, MD

Syllabus 220
Clinical Pharmacology for the Nephrologist—Suzanne K. Swan, MD,
Ali Olyaei, PharmD, and Domenic A. Sica, MD
Altered Pharmacokinetic Principles . . . . . . . . . . . . . . . . . . .220
Stepwise Approach to Dosimetry . . . . . . . . . . . . . . . . . . . . .222
Step 1: History and Physical Examination . . . . . . . . . . . .222
Step 2: Assess Renal Function . . . . . . . . . . . . . . . . . . . . .223
Step 3: Determine Loading Dose . . . . . . . . . . . . . . . . . . .224
Step 4: Determine Maintenance Dose . . . . . . . . . . . . . . .224
Step 5: Drug-Level Monitoring . . . . . . . . . . . . . . . . . . . .224
Drug Clearance in Dialysis . . . . . . . . . . . . . . . . . . . . . . . .225
Antimicrobial Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .227
Antibacterial Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . .227
Antifungal Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .228
Amphotericin B . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .228
Azoles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .229
Echinocandins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .230
Antiviral Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .230
Hepatitis B and C Antiviral Agents. . . . . . . . . . . . . . . .230
Anti-Influenzal Agents . . . . . . . . . . . . . . . . . . . . . . . . . .230
NephSAP
®
Volume 9, Number 4, July 2010

Antiretroviral Agents . . . . . . . . . . . . . . . . . . . . . . . . . . .230


Psychiatric Medications . . . . . . . . . . . . . . . . . . . . . . . . . . . . .233
Antidepressant Drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . .233
Lithium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .235
Anticonvulsant Drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .236
Older Anticonvulsant Drugs . . . . . . . . . . . . . . . . . . . . . . .236
Newer ACDs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .237
“Traditional” Medications, Food Contaminants, and
Others . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .238
Use of Extracorporeal Measures to Remove Drugs and
Chemicals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .238
Immunosuppressive Therapies . . . . . . . . . . . . . . . . . . . . . . .240
Induction Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .240
Basiliximab and Daclizumab . . . . . . . . . . . . . . . . . . . . .240
Antithymocyte Globulin Equine/Rabbit. . . . . . . . . . . . .241
Muronomab-CD3 (OKT-3). . . . . . . . . . . . . . . . . . . . . . .241
Alemtuzumab . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .242
Maintenance Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . .242
Calcineurin Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . .242
Tacrolimus (FK-506) . . . . . . . . . . . . . . . . . . . . . . . . .245
Generic Formulations of CNIs . . . . . . . . . . . . . . . . . .246
Azathioprine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .246
MPA Derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .247
TOR Inhibitors: Sirolimus and Everolimus . . . . . . . . . .248
Common Drug–Drug Interactions . . . . . . . . . . . . . . . . . . .248
Pharmacokinetic Drug Interactions . . . . . . . . . . . . . . . . . .249
Cardiovascular Medications . . . . . . . . . . . . . . . . . . . . . . . . .252
Antihypertensive Agents . . . . . . . . . . . . . . . . . . . . . . . . . .252
Peripheral ␣ Blockers . . . . . . . . . . . . . . . . . . . . . . . . . . .252
NephSAP
®
Volume 9, Number 4, July 2010

Central ␣ Agonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . .253


␤ Blockers. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .253
Calcium Channel Blockers . . . . . . . . . . . . . . . . . . . . . . .253
Angiotensin-Converting Enzyme Inhibitors. . . . . . . . . .254
Angiotensin Receptor Blockers . . . . . . . . . . . . . . . . . . .255
Direct Renin Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . .255
Antianginal Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .256
Cholesterol-Lowering Agents . . . . . . . . . . . . . . . . . . . . . .256
Triglyceride-Lowering Agents . . . . . . . . . . . . . . . . . . . . .257
Antiarrhythmic Agents . . . . . . . . . . . . . . . . . . . . . . . . . . .258
Diuretics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .259
Thiazide-Type Diuretics . . . . . . . . . . . . . . . . . . . . . . . . . .259
Loop Diuretics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .260
Potassium-Sparing Diuretics . . . . . . . . . . . . . . . . . . . . . . .260
Mannitol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .261
Gastrointestinal Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . .261
Antidiabetic Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .262
Analgesic Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .264
Acetaminophen . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .264
Opioid Analgesic Agents . . . . . . . . . . . . . . . . . . . . . . . . .264

CME Self-Assessment Questions . . . . . . . . . . . . . . . . . . . . . 266


Questions Linked to UpToDate in Green

Upcoming Issues
Renal Bone Disease, Disorders of Divalent Ions, and
Nephrolithiasis—
Kevin Martin, MBBCh, and Stanley Goldfarb, MD . . . .September 2010
NephSAP
®
Volume 9, Number 4, July 2010

End-Stage Renal Disease and Dialysis—


Rajiv Agarwal, MD, and Rajnish Mehrotra, MD . . . . . .November 2010

Geriatric Nephrology—
Dimitrios G. Oreopoulos, MD, PhD, Vanita Jassal, MD, Ann M. O’Hare, MD,
Mitchell H. Rosner, MD, Mark A. Swidler, MD, Jocelyn Wiggins, BM, BCh,
Mark E. Williams, MD, and Richard J. Glassock, MD . . . . .January 2011

Fluid, Electrolyte, and Acid-Base Disturbances—


Richard H. Sterns, MD, and Michael Emmett, MD . . . . . . .March 2011

Acute Kidney Injury and Critical Care Nephrology—


Patrick T. Murray, MD, and Kathleen D. Liu, MD . . . . . . . . .May 2011
NephSAP
®
Volume 9, Number 4, July 2010

The Editorial Board of NephSAP extends its sincere appreciation to the following reviewers. Their efforts and insights have helped to
improve the quality of this postgraduate education offering.
NephSAP Review Panel
Nihal Y. Abosaif, MBBCh Rajiv Dhamija, MD Pranay Kathuria, MD, FASN
St. James University Hospital Walk in Medical Care University of Oklahoma College of Medicine
Leeds, United Kingdom Artesia, CA Tulsa, OK
Georgi Abraham, MBBS Susan R. DiGiovanni, MD Quresh T. Khairullah, MD, FASN
Sri Ramachandra University Virginia Commonwealth University St. Clair Specialty Physicians
Medical Center Richmond, VA Detroit, MI
Chennai, India
Francis Dumler, MD Apurv Khanna, MD
Pablo H. Abrego, MD, FASN William Beaumont Hospital SUNY Upstate Medical University
Marshfield Clinic Royal Oak, MI Syracuse, NY
Wausau, WI
Mahmoud T. El-Khatib, MD, PhD, FASN Ramesh Khanna, MD
Anil K. Agarwal, MD, FASN University of Cincinnati Medical Center University of Missouri at Columbia
Ohio State University Medical Center Cincinnati, OH School of Medicine
Columbus, OH Columbia, MO
Lynda A. Frassetto, MD, FASN
Mustafa Ahmad, MD University of California at San Francisco Edgar V. Lerma, MD, FASN
King Fahad Medical City San Francisco, CA University of Illinois at Chicago
Riyadh, Saudi Arabia College of Medicine
Duvuru Geetha, MD
Chicago, IL
Jafar Al-Said, MD, FASN Johns Hopkins University
Bahrain Specialist Hospital Baltimore, MD Meyer D. Lifschitz, MD
Manama, Bahrain Shaare Zedek Medical Center
Carl S. Goldstein, MD
Jerusalem, Israel
Dante Amato-Martinez, MD, PhD Robert Wood Johnson Medical School
Universidad Nacional Autónoma de México New Brunswick, NJ Philippe S. Madhoun, MD
Tlalnepantla, Mexico Chu Charleroi
Nabil G. Guirguis, MD
Charleroi, Belgium
Anis U. Ansari, MD Kidney Dialysis and Transplant Group
Medical Associates Bridgeport, WV Jolanta Malyszko, MD, PhD, FASN
Clinton, IA Medical University
Pawan K. Gupta, MD
Bialystok, Poland
Akhtar Ashfaq, MD, FASN Altoona Regional Health System
North Shore University Hospital Altoona, PA Naveed N. Masani, MD
Great Neck, NY Winthrop University Hospital
Carsten Hafer, MD
Mineola, NY
Azra Bihorac, MD, FASN University of Hannover
University of Florida Hannover, Germany Hanna W. Mawad, MD, FASN
Gainesville, FL University of Kentucky Medical Center
Richard N. Hellman, MD
Lexington, KY
Mona B. Brake, MD Indiana University School of Medicine
Robert J. Dole VA Medical Center Indianapolis, IN Pascal Meier, MD, FASN
Wichita, KS Centre Hospitalier Universitaire Vaudois
Ekambaram M. Ilamathi, MD, FASN
Lausanne, Switzerland
Mauro Braun, MD Suffolk Nephrology Consultants
Cleveland Clinic Florida Stony Brook, NY Beckie Michael, DO, FASN
Weston, FL Marlton Nephrology and Hypertension
Viswanathan S. Iyer, MD, FASN
Marlton, NJ
Chokchai Chareandee, MD, FASN AKD-HTN LLC
Regions Hospital Harrisburg, PA Shahriar Moossavi, MD, PhD
Saint Paul, MN Wake Forest University Baptist
Bernard G. Jaar, MD
Medical Center
W. James Chon, MD, FASN Johns Hopkins Medical Institutions and
Winston-Salem, NC
University of Chicago Medical Center Nephrology Center of Maryland
Chicago, IL Baltimore, MD Scott R. Mullaney, MD
University of California at San Diego
Devasmita Choudhury, MD Avanelle V. Jack, MD
San Diego, CA
University of Texas Southwestern Louisiana State University Health
Medical School Sciences Center Quaid J. Nadri, MD, FASN
Dallas, TX New Orleans, LA King Faisal Specialist Hospital and
Research Center
Bulent Cuhaci, MD, FASN Sharon L. Karp, MD
Riiyadh, Saudi Arabia
Drexel University College of Medicine Indiana University School of Medicine
Philadelphia, PA Indianapolis, IN
Suzanne M. Norby, MD, FASN Karthik M. Ranganna, MD Robert J. Shay, MD, FASN
Mayo Clinic Drexel University College of Medicine East Georgia Kidney and
Rochester, MN Philadelphia, PA Hypertension Group
Augusta, GA
Michal Nowicki, MD Pawan K. Rao, MD, FASN
Medical University of Łódź St. Joseph’s Hospital Health Center Rolf A.K. Stahl, MD
Łódź, Poland Syracuse, NY University of Hamburg
Hamburg, Germany
Macaulay A. Onuigbo, MD, FASN Joel C. Reynolds, MD, FASN
Mayo Clinic Brooke Army Medical Center Harold M. Szerlip, MD, FASN
Eau Claire, WI San Antonio, TX Medical College of Georgia
Augusta, GA
Than N. Oo, MD Robert M.A. Richardson, MD
Nephrology Center University of Toronto Bekir Tanriover, MD
Kalamazoo, MI Toronto, ON, Canada Dialysis Nephrology Associates
Dallas, TX
Kevin P. O’Reilly, MD Bijan Roshan, MD
Columbus Nephrology, Inc. Joslin Diabetes Center Tushar J. Vachharajani, MD, FASN
Columbus, OH Harvard Medical School Wake Forest University
Boston, MA School of Medicine
Malvinder S. Parmar, MB, MS, FASN
Winston-Salem, NC
Northern Ontario School of Medicine Abinash C. Roy, MD
Timmins, ON, Canada University of Utah School of Medicine Allen W. Vander, MD, FASN
Saint George, UT Kidney Center of South Louisiana
Pairach Pintavorn, MD, FASN
Thibodaux, LA
East Georgia Kidney and Hypertension Mario F Rubin, MD
Augusta, GA Massachusetts General Hospital Luigi Vernaglione, MD
Boston, MA M. Giannuzzi Hospital
Paul H. Pronovost, MD, FASN
Manduria, Italy
Yale University School of Medicine Ehab R. Saad, MD, FASN
Waterbury, CA Medical College of Wisconsin Shefali Vyas, MD
Milwaukee, WI Saint Barnabas Medical Center
Mohammad A. Quasem, MD, FASN
Livingston, NJ
State University of New York Mohammad G. Saklayen, MD
Binghamton, NY Wright State University Medical School Alexander Woywodt, MD, FASN
Dayton, OH Lancashire Teaching Hospitals NHS
Wajeh Y. Qunibi, MD
Foundation Trust
University of Texas Health Sciences Center Ramesh Saxena, MD, PhD
Preston, United Kingdom
San Antonio, TX University of Texas Southwestern
Medical Center
Venkat Ramanathan, MD, FASN
Dallas, TX
Baylor College of Medicine
Houston, TX Gaurang M. Shah, MD
Long Beach VA Healthcare System
Long Beach, CA
NephSAP
®
Volume 9, Number 4, July 2010

Program Mission and Objectives


The mission of the Nephrology Self-Assessment Program (NephSAP) is to regularly provide a vehicle that will be useful for clinical
nephrologists who seek to renew and refresh their clinical knowledge and diagnostic and therapeutic skills. This Journal consists of a
series of challenging, clinically oriented questions based on case vignettes, a detailed Syllabus that reviews recent publications,
and an Editorial on an important and evolving topic. Taken together, these parts should assist individual clinicians under-
taking a rigorous self-assessment of their strengths and weaknesses in the broad domain of nephrology.

Accreditation and Credit Designation


The American Society of Nephrology is accredited by the Accreditation Council for Continuing Medical Education to provide con-
tinuing medical education for physicians.
The ASN designates this educational activity for a maximum of 8.0 AMA PRA Category 1 Credits™. Physicians should only claim
credit commensurate with the extent of their participation in the activity.

Continuing Medical Education (CME) Information


CME Credit: 8.0 AMA PRA Category 1 Credits™
Date of Original Release: July 2010
Examination Available Online: on or before Friday, July 9, 2010
Audio Files Available: No audio files will be available for this issue.
CME Credit Eligible Through: June 30, 2011
Answers: Correct answers with explanations will be posted on the ASN website in July 2011 when the issue is archived.
UpToDate Links Active: July and August 2010
Core Nephrology question links active: There are no core questions for this issue.
Target Audience: Nephrology Board and recertification candidates, practicing nephrologists, and internists.
Method of Participation:
● Read the syllabus that is supplemented by original articles in the reference lists, and complete the online self-assessment
examination.
● Examinations are available online only after the first week of the publication month. There is no fee. Each participant is
allowed two attempts to pass the examination (⬎75% correct).
● Your score and a list of question/s (by number) answered incorrectly can be printed immediately.
● Your CME certificate can be printed immediately after passing the examination.
● Answers and explanations are provided ONLY with a passing score on the first or second attempt.
● Your ASN transcript will be updated in 6 to 8 weeks after passing the examination.
Instructions to access the Online CME Center to take the examination and complete the evaluation:
● Login at ASN’s website: www.asn-online.org
● Click the “CME” tab at the top of the homepage
● In the left-hand column click “Online CME Center”
● Scroll down; in the center of the page click the computer screen image to “Visit the Online CME Center”
● Once you’re in the Center, click the “NephSAP” banner in the center of the page
● Select a topic and click on “Enter Activity” to the right
● Read through the instructions, and then click on “Continue” at the bottom of the page
● Click on “Examination Questions/Evaluations” to begin
● Your score and a list of question numbers answered incorrectly can be printed immediately
● Follow the prompts to retake the examination if you failed, or print your certificate and the correct answers if you passed
● You can save and finish the exam to complete at another time
● You can retake the exam at any time; each participant is allowed two attempts
NephSAP
®
Volume 9, Number 4, July 2010

Instructions to Obtain American Board of Internal Medicine (ABIM) Maintenance of Certification


(MOC) Points:
Each issue of NephSAP provides 10 MOC points. Respondents must meet the following criteria:
● Be certified by ABIM in internal medicine and/or nephrology and must be enrolled in the ABIM–MOC program
via the ABIM website (www.abim.org).
● Pass the self-assessment examination within the timeframe specified in this issue of NephSAP.
● Designate the issue for MOC points by clicking on the MOC link on the CME certificate page after passing the examination.
You will be leaving the ASN-CECity site and transferring the information directly to the ABIM in real-time.
● Provide your ABIM Certificate ID number and your date of birth.
● You will receive a confirmation message from the ABIM indicating the receipt of your information.
MOC points will be applied to only those ABIM candidates who have enrolled in the program. It is your responsibility to complete
the ABIM MOC enrollment process.

Instructions to access the ASN website, NephSAP, and the UpToDate link
Compatible Browser: The ASN website (asn-online.org) has been formatted for cross-browser functionality, and should
display correctly in all modern web browsers. We recommend that you use Internet Explorer.
Monitor Settings: The ASN website was designed to be viewed in a 1024 ⫻ 768 or higher resolution.
Technical Support: If you are having difficulty viewing any of the pages, please refer to the ASN technical support page
for possible solutions. If you continue having problems, contact Hal Nesbitt at hnesbitt@asn-online.org.
UpToDate provides an additional source of information that should help you answer up to 5 selected NephSAP
questions from each issue. The link is free and will remain active for the first 60 days after publication for each
issue.
● On the ASN home page, double click on the NephSAP link (NephSAP cover) on the bottom side of the page.
● On the NephSAP page, click on the UpToDate button on the left hand side to access the current links.
NephSAP
®
Volume 9, Number 4, July 2010

Disclosure Information
The ASN is responsible for identifying and resolving all conflicts of interest prior to presenting any educational activity to learners to ensure
that ASN CME activities promote quality and safety, are effective in improving medical practice, are based on valid contents, and are inde-
pendent of the control from commercial interests and free of commercial bias. All faculty are instructed to provide balanced, scientifically
rigorous and evidence-based presentations. In accordance with the disclosure policies of the Accreditation Council for Continuing Medical
Education (ACCME) as well as guidelines of the Food and Drug Administration (FDA), individuals who are in a position to control the con-
tent of an educational activity are required to disclose relationships with a commercial interest if (a) the relation is financial and occurred
within the past 12 months; and (b) the individual had the opportunity to affect the content of continuing medical education with regard to that
commercial interest. For this purpose, ASN consider the relationships of the person involved in the CME activity to include financial relation-
ships of a spouse or partner. Peer reviewers are asked to abstain from reviewing topics if they have a conflict of interest. Disclosure informa-
tion is made available to learners prior to the start of any ASN educational activity.
Agarwal, Rajiv—Research funding: Abbott; Consultant/scientific advisor: Rockwell Medical, Watson Pharma; Honoraria: Abbott, Astra-
Zeneca, Merck
Berns, Jeffrey S.—Consultant: Amgen; Research funding: NxStage; Advisory board: Affymax, NAAC
Cohen, David J.—Research funding: Life Cycle Pharma, Novartis, Roche, Wyeth; Honoraria: Bristol-Myers-Squibb, Novartis, Roche
Emmett, Michael—Honoraria: ABIM, Albert Einstein Medical Center, American Renal Associates, Best Doctors, Braintree Labs,
Brown University, Fresenius, Partners-Boston, Renal Ventures, Shire, Temple Medical, TIPS; Editorial board membership:
American Journal of Cardiology, Baylor Proceedings
Fishbane, Steven—Consultant: Abbott, Affymax, AMAG, Genzyme, Watson; Research funding: Affymax, Biogen Idec, Genentech,
Takeda; Honoraria: Abbott, Affymax, AMAG, Takeda, Watson; Expert testimony: Roche; Advisory board: Affymax, Genzyme,
Takeda, Watson
Fuchs, Elissa (Medical Editor)—none
Glassock, Richard J.—Consultant: Bio-Marin, Bristol Myers Squibb, FibroGen, Genentech, Gilead, Lighthouse Learning, Novartis,
Quest Diagnostics/Nichols Institute, Science Partners, QuestCor, Vifor (Aspreva) Pharmaceutical, Wyeth; Ownership interests:
LaJolla Pharmaceutical, Reata Inc.; Honoraria: Various medical schools—Columbia University, University of South Florida, USC,
Cedars Sinai Medical Center, Lupus Foundation of America; Membership board of directors/scientific advisor: Los Angeles
Biomedical Institute, University Kidney Research Associates, Vifor (Aspreva) Pharmaceutical, Wyeth; Editorial board: UpToDate;
Royalties: Lippincott, Williams and Wilkins, Oxford University Press
Goldfarb, Stanley—Consultant: Lupitold, Omeros; Ownership interests: Polymedix; Honoraria: GE Healthcare, Fresenius; Expert
testimony: Bayer; Editorial board: Journal of Clinical Investigation
Martin, Kevin J.—Research funding/honoraria: Abbott; Advisory board: Abbott, Cytochroma
Mehrotra, Rajnish—Research funding: Amgen, Baxter, Shire; Consultant: Novartis; Honoraria: AMAG, Baxter Healthcare, Shire
Murray, Patrick T.—Employment: spouse, Merck, Sharpe, & Dohme (Europe); Consultant/honoraria/research funding/
scientific advisor: Abbott Laboratories (USA), Argutus Medical (UK), FAST Diagnostics (USA); NxStage Medical (USA).
Nachman, Patrick H.—Honoraria: QuestCor; Multicenter clinical trial participation: Otsuka
Palevsky, Paul M.—Consultant: Nephrion; Advisory board/membership: Member, Board of Directors, Renal Physicians Association;
President ESRD Network 4
Sterns, Richard H.—Consultant: Astellas, Otsuka; Honoraria: Astellas, Otsuka; UpToDate; Scientific advisor: UpToDate
Textor, Stephen C.—Editorial board membership: Journal of the American Society of Nephrology, Clinical Journal of the American
Society of Nephrology
Townsend, Raymond T.—Consultant: Daiichi-Sankyo, GlaxoSmithKline, Merck, Nicox, Novartis, Roche; Research funding: Novartis;
Honoraria: American Society of Hypertension, National Kidney Foundation
Vella, John P.—none
Guest Co-Editors:
Swan, Suzanne K.—none
Olyaei, Ali—Consultant: Drug Safety and Research Institute; Research funding/honoraria: Astella, Novartis, Roche
Sica, Domenic A.—Consultant: Bayer, Novartis, Takeda; Research funding: CVRx, GlaxoSmithKline, Novartis, Takeda; Honoraria:
Boehringer Ingelheim, Daiichi Sankyo, Forest, Ligand, Merck, Novartis, Sanofi/Bristol-Myers Squibb, Takeda; Scientific advisor/
member editorial board: Section editor-Journal of Clinical Hypertension and Cardiology Reviews, Merit Review committee for
Veteran’s Administration, NIH study section, Sun Tech
Editorial:
Perazella, Mark A.—Membership: ABIM Nephrology Subspecialty Board

Commercial Support
There is no commercial support for this issue.
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

Editorial
Pharmacology and Kidney Disease: Lessons Learned from the
Gadolinium–Nephrogenic Systemic Fibrosis Story
Mark A. Perazella, MD
Section of Nephrology, Department of Medicine, Yale University School of Medicine,
New Haven, Connecticut

Very infrequently does a new disease suddenly oxidation state in aqueous solutions and has an ionic
appear in a group of patients who have been closely radius nearly equal to divalent calcium (Ca2⫹). Be-
observed for the past 30 to 40 years. Nonetheless, this cause of this characteristic, free ionic gadolinium
occurred in patients with chronic kidney disease (Gd3⫹) competes with Ca2⫹ in biologic systems (3).
(CKD) and ESRD when nephrogenic systemic fibrosis Because Gd3⫹ has a higher binding affinity than Ca2⫹,
(NSF) caught the nephrology community by surprise it readily reduces neuromuscular transmission by ob-
with its initial identification in 1997 and its subsequent structing Ca2⫹ channels in muscle and nerve tissue
formal description as a unique entity in 2001 (1). cells and alters enzyme kinetics (3). In addition, the
During the subsequent several years, the cause of NSF lanthanides enhance the polymerization rate of colla-
eluded diagnosis, but it was clear that it occurred only gen and mediate fibrillogenesis by direct binding to
in patients with kidney disease. In 2006, a break- the collagen helix (4). These effects suggest potential
through occurred as the causative agent: Gadolinium- for toxicity.
based contrast agents (GBCAs) were identified and For safe use of the contrast-enhancing benefits of
subsequently verified in numerous studies (2). Gd3⫹ with magnetic resonance imaging (MRI), the
As with any newly recognized disease or com- metal must be sequestered by an organic ligand (3).
plication in medicine, an adverse drug effect must be Chelation of Gd3⫹ to a nontoxic ligand allows forma-
suspected, and, in general, it is reasonable to assume tion of a stable complex and renders the metal bio-
that “it is a drug until proven otherwise.” In this case, chemically inert (3). A few important points about
a drug in the guise of a magnetic resonance contrast– Gd3⫹–ligand complex stability are reviewed to permit
enhancing imaging agent was the culprit. As we look an understanding of GBCA differences in causing
back and ponder this new disease called NSF, the NSF. In general, there are two major categories of
question that lingers for all of us is whether toxicity ligand, linear and macrocyclic, which are based on
was a predictable complication of the GBCAs. When biochemical structure. Charge (ionic versus nonionic)
faced with such a question, we must return to basics allows further subclassification of the GBCAs.
and examine whether the drug chemistry and/or its The “tightness” with which a ligand binds Gd3⫹, or
pharmacokinetics in patients with kidney disease sug- “complex stability,” is determined primarily by the
gest potential for toxicity. amine characteristics of the ligand. Linear ligands have
amines with amide-containing side chains, whereas mac-
Gadolinium Chemistry rocyclic ligands have acetate side chains, which are more
A quick look at the chemistry of gadolinium and basic and form more stable and presumably safer com-
the lanthanides is a worthwhile starting point. Gado- plexes (3). For measurement of complex stability, a
linium has paramagnetic properties that disturb relax- number of yardsticks are used, including the ther-
ation of water protons and by shortening relaxation modynamic stability constant, conditional stability
times increases signal intensity, making it an excellent constant, and dissociation kinetics. In a test tube,
contrast agent. Like all lanthanides, it exists in the ⫹3 the thermodynamic and conditional stability con-
213
214 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

stants are reflective of ligand affinity to Gd3⫹; Rats that were exposed to GBCAs had tissue
3⫹
however, in biologic media such as human blood, in Gd concentrations that were highest with linear-
contrast to the in vitro system, additional competi- based agents and were approximately 30-fold lower
tors for ligand binding are present (3). These in- with two macrocyclic agents (6). Long-term retention
clude zinc, copper, and iron, which are capable of of linear GBCAs was noted within skin, whereas three
forming very stable complexes with the organic different macrocyclic GBCAs had unmeasurable con-
ligand. Also, Gd3⫹ has high affinity for endogenous centrations (7). Partially nephrectomized rats that were
anions such as phosphate, carbonate, and citrate, exposed to various linear-based GBCAs developed
which compete with the ligand. significant Gd3⫹ concentrations in various tissues as
In this situation, the rate of complex dissociation compared with saline and chelate-only controls (8).
or kinetic stability is more reflective of actual stability Finally, 5/6-nephrectomized rats had skin concentra-
of the GBCA in the bloodstream. The more kinetically tions at day 168 that were highest with linear GBCAs,
inert the complex, the less likely it will dissociate and which were approximately 500 times higher than mac-
participate in “transmetallation.” Transmetallation rocyclic GBCAs (9).
(Figure 1) entails Gd3⫹ dissociation from its ligand, In humans, Gd3⫹ is noted within tissues after
whereby the ligand binds an endogenous metal such as GBCA exposure. Bone concentrations were higher
zinc, copper, or iron and Gd3⫹ binds an endogenous with linear- versus macrocyclic-based agents. For ex-
substance such as phosphate, citrate, or carbonate (5). ample, after 0.1-mmol/kg dosing with GBCA in
This process is more likely to occur the longer the healthy individuals, Gd3⫹ bone concentrations were
GBCA resides in the bloodstream, which as is dis- nearly four times higher with a linear agent as com-
cussed later is very relevant to patients with underly- pared with macrocyclic agents (10). Furthermore, pa-
ing kidney disease. Thus, the safest GBCA is one that tients with NSF had Gd3⫹ tissue concentrations that
dissociates the slowest— essentially one that is kinet- are 35 to 150 times higher than those in normal control
ically inert. subjects who were exposed to linear agent gadodia-
On the basis of this information, we can make mide (11,12).
predictions about the various GBCAs as it relates to
complex kinetic stability and risk for transmetallation. Gadolinium-Based Contrast Pharmacology
Despite data that reflect reasonably good thermody- The toxicity of most drugs relates primarily to
namic and conditional stability constants with linear two factors: (1) innate drug toxicity and (2) overall
GBCAs, they are not as kinetically stable as macro- host exposure. We know that Gd3⫹ has toxic potential.
cyclic agents and will dissociate more readily in a The exposure issue is addressed by examining the
biological system. In fact, animal studies and obser- pharmacokinetics of these agents. Area under the
vations in humans who were exposed to GBCA sup- curve as a measure of exposure is primarily driven by
port these data. dosage, distribution half-life (T ⁄ ), and elimination
12

Gd3+
Fe3+
Gd3+ Gd3+
Ca2+
C Gd3+
2+ 3+
Zn Gd
Gd3+ Fe3+
F
Cu2+ 3+
Gd (CH3)NHCO CONH(CH3)
(CH3)NHCO CONH(CH3) N N N
N N N HOOC COOH
HOOC COOH Ca2+
C

(CH3)NHCO CONH(CH3)
N N N
HOOC COOH
Figure 1. Transmetallation of injected GBCA. A linear GBCA binds Gd3⫹ less tightly than other ligands (macrocyclic),
allowing endogenous cations such as copper (Cu2⫹), iron (Fe3⫹), zinc (Zn2⫹), and Ca2⫹ to compete with Gd3⫹ for ligand
binding. This allows free Gd3⫹ to be released into the bloodstream, where it may bind other anions such as phosphate.
Abbreviations: GBCA, gadolinium-based contrast agent. Reprinted from reference 5, with permission.
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 215

T ⁄ . Injected GBCAs are rapidly distributed into the


12 a result of albumin binding and absence of hepatic
extracellular space and quickly equilibrate between the clearance (14).
plasma and interstitial compartments (Figure 2), con- The Vd for ECF GBCAs is unchanged in patients
sistent with a two-compartment model (13). They do with kidney disease; however, renal clearances are
not undergo biotransformation, are not protein bound, much lower in stages 3 and 4 CKD than in normal
and have a small volume of distribution (Vd; approx- individuals. As a result, the terminal T ⁄ is prolonged
12

imately 0.26 L/kg). Although the bulk of available in patients with stage 3 (4 to 8 hours) and stage 4 (18
GBCAs are extracellular fluid (ECF) agents, some are to 34 hours) CKD (13,14), increasing total agent
ECF agents that also enter liver cells, whereas others excretion time from 24 hours to ⱖ1 week. Agents with
are albumin bound and limited to the plasma compart- a component of liver clearance have a terminal T ⁄ of
12

ment (Vd approximately 0.15 L/kg) (14). 6.1 and 9.5 hours with stages 3 and 4 CKD, respec-
The ECF GBCAs are eliminated unchanged by tively (14). Gadoxetate has a terminal T ⁄ of 12 hours
12

the kidneys via glomerular filtration without any tu- in patients with ESRD. It is also not well appreciated
bular secretion (Figure 2). Renal clearance approxi- that CKD is associated with reduced hepatic drug
mates creatinine clearance in normal individuals. They metabolism, potentially limiting hepatic clearance of
possess a terminal T ⁄ of approximately 1.3 to 1.6
12 GBCAs that also use this excretion pathway. For
hours, and ⬎95% of an injected dose is eliminated gadofosveset, stages 3 and 4 CKD, respectively, are
within 24 hours (13). Hence, kidney function is im- associated with two- and threefold increases in termi-
portant in determining overall terminal T ⁄ and drug
12 nal T ⁄ above that seen in normal individuals (14).
12

exposure. Certain GBCAs have a component of he- Pharmacokinetic data of GBCAs predict that
patic clearance; 2 to 4% for gadobenate and 50% for they can be removed relatively efficiently by hemodi-
gadoxetate (14). These agents are associated with a alysis. On average, the T ⁄ of GBCAs in nondialysis
12

terminal T ⁄ in normal individuals of 1.5 to 2.0 hours


12 patients with ESRD is 34.3 hours but decreases sig-
and 1.0 hour, respectively. In contrast, terminal T ⁄ for12 nificantly to 2.6 hours with hemodialysis (13). GBCA
the plasma-limited agent gadofosveset is 18.5 hours as serum elimination using hemodialysis is approxi-
mately 74% with one treatment, 92% with two treat-
ments, and 99% after three treatments (13). In con-
trast, low-dosage peritoneal dialysis (8 to 10 L/d) is
ineffective in GBCA removal, with T ⁄ of approxi-
12
GBCA
mately 53 hours (13). In general, 6 to 8 hours of
hemodialysis using high-flux membranes would pre-
dictably remove a significant amount of GBCA.
GBCA GBCA
GBCA Imaging in Kidney Disease
Intravascular Interstitial
On the basis of the chemistry and pharmacology
space space of the available GBCAs, perhaps we should have been
more vigilant about the potential for adverse effects in
Extra-cellular space
patients with kidney disease; however, we were reas-
GBCA sured by data from GBCA-exposed normal individuals
that supported safety. Food and Drug Administration
approval for the five US GBCAs between 1988 and
2004 was based on an excellent safety profile from
combined premarketing studies on ⬎3000 patients.
GBCA
The most common adverse effect was a mild allergic
reaction that resulted in rash, sweating, itching, hives,
Figure 2. Pharmacokinetics of injected GBCA. GBCA is and facial swelling. Only rarely was the reaction
distributed within the extracellular space (two-compartment
model) and is excreted primarily by the kidneys for all
severe with potential for fatality. No other notable
agents. A few agents are excreted in variable amounts by the acute or chronic adverse effects were recognized. In
liver. fact, millions of GBCA doses have been used safely in
216 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

patients with normal kidney function—again very re- the literature suggests that a threshold level of kidney
assuring. function must be crossed for NSF to develop after
Studies were also undertaken of patients with GBCA exposure. Dialysis-dependent ESRD, stage 5
various levels of CKD. Unfortunately, few patients CKD, and AKI maintain the highest risk for NSF.
who either had advanced CKD or dialysis were in- Nearly 80% are dialysis-dependent patients with
cluded. Most studies were small and had short-term ESRD. When one considers the much larger number
follow-up. The only adverse effect, which was not of patients with stage 4 CKD relative to those with
universally noted in all studies, seemed to be modest stage 5 and ESRD, risk in this group is minimal
risk for nephrotoxicity, especially with higher GBCA because only a very small number of patients with
dosages in high-risk patients (15). The perceived stage 4 CKD have developed NSF. Notably, no cases
safety of these agents led to increased use of GBCA in stages 1 through 3 CKD have been confirmed in the
MRI in the early to mid-1990s. In particular, renal and published literature. In fact, several studies of patients
peripheral vessels, the central nervous system, and the who had stages 1 through 4 CKD and were exposed to
liver were widely imaged in patients with acute kidney GBCAs did not note NSF as a complication (16 –18).
injury (AKI), CKD, and dialysis-dependent ESRD. In the Halt Progression of Polycystic Kidney Disease
Furthermore, contrast volumes two to three times the (HALT-PKD) study and Consortium of Radiologic
approved dosage (0.1 mmol/kg) were commonly used Imaging Study of PKD (CRISP), patients with auto-
for angiography. The initial recognition of NSF as a somal polycystic kidney disease and stages 1 through
new and unusual fibrosing disorder in patients with 3 CKD in underwent 1111 GBCA exposures with no
CKD in 1997 possesses an eerie temporal association occurrence of NSF (17). A total of 592 patients who
with the sudden spike in GBCA exposure in this had stages 3 through 4 CKD and were exposed to
group. That Gd3⫹ has collagen fibrillogenesis proper- GBCA also did not develop NSF (16). Eighty-eight
ties (4) makes this observation more relevant. patients who had stages 1 through 4 CKD and under-
went 94 GBCA exposures, including 38 patients with
NSF and Kidney Disease: Who Is Most at Risk? stage 4 CKD, did not develop NSF (18). Finally, none
Because NSF is a devastating and essentially of 2053 patients (stage 3, 44.7%; stage 4, 23.9%; stage
untreatable disease, it is our responsibility as physi- 5, 5.7%) who were exposed to 2278 GBCA MRIs
cians to reduce its occurrence. We must also remem- (various formulations) developed NSF during an av-
ber that our very strong emotional reaction to avoid all erage follow-up period of nearly 29 months (19). It
GBCA exposure in patients with kidney disease has thus seems that advanced kidney disease with very low
potential untoward consequences. Our patients often GFR levels portends the highest risk, whereas stage 4
have disorders that require accurate imaging to allow CKD seems to be very low risk and stages 1 through
rapid diagnosis and intervention. Missing lesions such 3 have negligible risk.
as liver cancer or serious brain pathology with subop- One is left to wonder why NSF occurs so infre-
timal imaging choices will likely lead to adverse quently compared with other complications that affect
outcomes. Also, exposing such patients to iodinated patients with kidney disease. The explanation almost
radiocontrast studies in lieu of GBCA has several certainly lies with the fact that other patient-related
possible complications. These include an increased factors must coexist for NSF to develop, the proverbial
number of potentially life-threatening allergic reac- “perfect storm.” Advanced kidney disease and GBCA
tions, significant radiation exposure, and nephrotoxic- exposure are necessary but not sufficient in and of
ity. The last complication is of particular concern themselves. A number of putative co-factors have
because there are data to support that AKI after radio- been previously reviewed (Table 1) and include vari-
contrast may be associated with increased morbidity ous systemic and metabolic disorders and certain ther-
and mortality, whereas loss of residual renal function apies (20). Linear-based GBCAs themselves induce
in dialysis patients has untoward consequences. Thus, inflammation, perhaps contributing further to patient
it is incumbent on the nephrology community to iden- risk (20). Inflammatory states may facilitate profi-
tify patients who are at highest risk and address their brotic cytokine and chemokine synthesis, which would
imaging needs in a rational, evidence-based manner. attract profibrotic cells to tissues that contain Gd3⫹
In addition to certain formulations of GBCA (linear), and increase collagen formation.
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 217

Table 1. Putative co-factors for NSF result of increased Gd3⫹ tissue exposure in patients
Proinflammatory states with CKD.
infection
connective tissue diseases
major surgery How Should We Image High-Risk Patients with
Hypercoagulable states CKD?
Vascular injury Because patients with CKD have numerous
Liver failure comorbidities that necessitate imaging tests, we
hepatorenal syndrome must make rational choices that are guided by pub-
liver transplantation lished data. The natural tendency is to avoid GBCA
Metabolic abnormalities
use in all patients with any form of kidney disease,
hyperphosphatemia
hypercalcemia but this approach has consequences, as described
metabolic acidosis previously. On the basis of what we currently know,
iron overload the following statements on patient risk for NSF can
Therapeutic agents be made:
high-dosage ESA
intravenous iron Y Patients with stages 1 through 3 CKD do not seem to
ESA, erythropoiesis-stimulating agent. maintain risk for NSF after GBCA exposure.
Y Patients with stage 4 CKD maintain modest risk, per-
haps increased when other factors (e.g., inflammation,
The vast majority of clinical observations pub-
metabolic abnormalities) are present.
lished on NSF after GBCA exposure support a rela- Y Patients with stage 5 CKD and AKI are at high risk,
tionship to dosage, whether single high-dosage or with dialysis-dependent patients with ESRD at highest
higher cumulative dosage. Furthermore, at one center, risk.
a protocol change that switched use from gadodiamide
to gadobenate, an agent that allows excellent imaging In my view, imaging options for patients who
with use of lower dosages (0.2 versus 0.1 mmol/kg), are defined as being at high risk for NSF should be
was associated with a reduction in NSF incidence from approached logically. First, use a non-GBCA–
2.6 to 0.0% (21). These data support high-dosage requiring technology that is safe and suitably di-
administration as a risk factor for NSF, likely as a agnostic (Table 2). When imaging with a GBCA

Table 2. Non– gadolinium-based contrast agent–requiring imaging technologies


Ultrasonography
grayscale ultrasonography provides information about organ size and echogenicity
color, power, and spectral Doppler ultrasonography may be useful for renal artery stenosis and other vascular disease;
this modality is highly operator dependent with only a few centers with expertise
contrast with microbubbles views liver lesions very well but is not FDA approved or available in the United States and
has a black box warning for patients with “cardiovascular instability”
CT scan
non–radiocontrast-enhanced is an option for some forms of imaging
radiocontrast-enhanced CT scan is an option for many (but not all) imaging needs
risk of radiation exposure, contrast allergy, and nephrotoxicity (AKI and loss of residual renal function)
MRA
contrast enhancement with ultrasmall superparamagnetic iron oxide (ferumoxytol) offers excellent imaging of various
organs such as brain and vasculature (aorta, carotid, peripheral arteries)
non-enhanced MRA techniques can image renal vessels and other vasculature very well without contrast
time of flight
arterial spin labeling
perfusion contrast angiography
steady-state free precession
nonenhanced MRI technique (diffusion-weighted) can image liver pathology very well
CT, computed tomography; FDA, Food and Drug Administration; MRA, magnetic resonance angiography.
218 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

is required, the following approach should be cause what is safe in patients with normal kidney
considered: function may not be in this group.
Y Use only macrocyclic GBCAs; agents with hepatic
References
clearance should be studied further to document safety. 1. Cowper SE, Su LD, Bhawan J, Robin HS, LeBoit PE: Nephrogenic
Y Use the lowest dosage of GBCA required to provide fibrosing dermopathy. Am J Dermatopathol 23: 383–393, 2001
quality images. 2. Grobner T: Gadolinium: A specific trigger for the development of
Y Avoid intravenous iron and high-dosage erythropoie- nephrogenic fibrosing dermopathy and nephrogenic systemic fibro-
sis? Nephrol Dial Transplant 21: 1104 –1108, 2006
sis-stimulating agents before and after the anticipated
3. Sherry AD, Caravan P, Lenkinski RE: Primer on gadolinium chem-
exposure. istry. J Magn Reson Imaging 30: 1240 –1248, 2009
Y Optimize calcium, phosphate, and acid-base balance 4. Stratta P, Canavese C, Quaglia M, Fenoglio R: Gadolinium-associ-
before the anticipated exposure. ated nephrogenic systemic fibrosis in patients with renal failure: The
Y Consider arranging hemodialysis to follow GBCA ex- need for an interdisciplinary helping network. Rheumatology (Ox-
ford) 49: 821– 823, 2010
posure for patients who are currently on this modality.
5. Perazella MA: How should nephrologists approach gadolinium-based
The recommendation of hemodialysis after GBCA contrast imaging in patients with kidney disease? Clin J Am Soc
Nephrol 3: 649 – 651, 2009
exposure is based primarily on pharmacokinetic data and 6. Sieber MA, Lengsfeld P, Frenzel T, Golfier S, Schmitt-Willich H,
the hypothetical benefit of removing this agent from the Siegmund F, Walter J, Weinmann HJ, Pietsch H: Preclinical inves-
bloodstream, thereby decreasing tissue exposure. Retro- tigation to compare different gadolinium-based contrast agents re-
garding their propensity to release gadolinium in vivo and to trigger
spective studies reveal conflicting results showing no nephrogenic systemic fibrosis-like lesions. Eur Radiol 18: 2164 –
preventive benefit of hemodialysis (22) or a possible 2173, 2008
reduction in NSF occurrence (23) when performed on the 7. Pietsch H, Lengsfeld P, Jost G, Frenzel T, Hütter J, Sieber MA:
Long-term retention of gadolinium in the skin of rodents following
day of exposure. This may relate to differences in the the administration of gadolinium-based contrast agents. Eur Radiol
actual timing of dialysis after exposure (sooner versus 19: 1417–1424, 2009
8. Grant D, Johnsen H, Juelsrud A, Lovhaug D: Effects of gadolinium
later). In reality, this issue has not been rigorously stud-
contrast agents in naïve and nephrectomized rats: Relevance to
ied, and, in my opinion, the ability to perform hemodi- nephrogenic systemic fibrosis. Acta Radiol 50: 156 –169, 2009
alysis after exposure in and of itself does not justify 9. Pietsch H, Lengsfeld P, Steger-Hartmann T, Löwe A, Frenzel T, Hütter J,
Sieber MA: Impact of renal impairment on long-term retention of gadolin-
GBCA administration to a high-risk patient. Rather, this ium in the rodent skin following the administration of gadolinium-based
modality should be considered for use, ideally within 2 to contrast agents. Invest Radiol 44: 226–233, 2009
3 hours of exposure and on consecutive days, in patients 10. White GW, Gibby WA, Tweedle MF: Comparison of Gd(DTPA-
BMA) (Omniscan) versus Gd(HP-DO3A) (ProHance) relative to
who are already undergoing short- or long-term mainte- gadolinium retention in human bone tissue by inductively coupled
nance hemodialysis. plasma mass spectroscopy. Invest Radiol 41: 272–278, 2006
11. Boyd AC, Zic JA, Abraham JL: Gadolinium deposition in nephro-
genic fibrosing dermopathy. J Am Acad Dermatol 56: 27–30, 2007
Conclusions 12. High WA, Eng M, Ayers RA, Cowper SE: Gadolinium is quantifiable
within the tissue of patients with nephrogenic systemic fibrosis. J Am
The imaging needs of patients with kidney
Acad Dermatol 56: 1–2, 2007
disease are significant, but as we have learned with 13. Perazella MA, Rodby RA: Gadolinium use in patients with kidney
iodinated radiocontrast, toxicity is an important ad- disease: A cause for concern. Semin Dial 20: 179 –185, 2007
14. Aime S, Caravan P: Biodistribution of gadolinium-based contrast
verse effect of GBCAs. Although nephrotoxicity is
agents, including gadolinium deposition. J Magn Reson Imaging 30:
minor with these agents, systemic toxicity can be 1259 –1267, 2009
substantial, as seen with the devastating illness 15. Perazella MA: Current status of gadolinium toxicity in patients with
kidney disease. Clin J Am Soc Nephrol 4: 461– 469, 2009
NSF. Focusing on GBCA agents as “drugs” reveals 16. Collidge TA, Thomson PC, Mark PB, Traynor JP, Jardine AG,
that, on the basis of the combination of the known Morris ST, Simpson K, Roditi GH: Gadolinium-enhanced MR im-
toxicity of free Gd3⫹ (Ca2⫹ channel blockade, en- aging and nephrogenic systemic fibrosis: Retrospective study of a
renal replacement therapy cohort. Radiology 245: 168 –175, 2007
hanced fibrillogenesis), the behavior of these agents 17. Chapman A, Grantham JJ, Torres VE, Guay-Woodford LM, Braun
in biologic systems (various chelate stability prop- W, Rahbari-Oskovi F, Kelleher C, Schrier RW, Perrone R, Steinman
erties), and their altered pharmacokinetics in pa- T, Miskulin D, Meyers C, Bost J, Miller P, Bae KT: Absence of NSF
following gadolinium exposure in ADPKD individuals with stable
tients with reduced GFR (increased T ⁄ ), one can
1 2
CKD [Abstract]. J Am Soc Nephrol 18: 837A, 2007
predict that some of these agents are potentially 18. Rydahl C, Thomsen HS, Marckmann P: High prevalence of nephro-
hazardous. NSF has taught us that we must bear in genic systemic fibrosis in chronic renal failure patients exposed to
gadodiamide, a gadolinium-containing magnetic resonance contrast
mind all of these issues when we use any drug or agent. Invest Radiol 43: 141–144, 2008
imaging agent in patients with kidney disease, be- 19. Chrysochou C, Power A, Shurrab AE, Husain S, Moser S, Lay J,
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 219

Salama AD, Kalra PA: Low risk for nephrogenic systemic fibrosis in ing gadolinium contrast-enhanced MRI protocols. J Magn Reson
nondialysis patients who have chronic kidney disease and are inves- Imaging 31: 440 – 446, 2010
tigated with gadolinium-enhanced magnetic resonance imaging. Clin 22. Broome DR, Girguis MS, Baron PW, Cotrell AC, Kjellin I, Kirk GA:
J Am Soc Nephrol 5: 484 – 489, 2010 Gadodiamide-associated nephrogenic systemic fibrosis: Why radiol-
20. Perazella MA: Advanced kidney disease, gadolinium and nephro- ogists should be concerned. AJR Am J Roentgenol 188: 586 –592,
genic systemic fibrosis: The perfect storm. Curr Opin Nephrol 2007
Hypertens 18: 519 –525, 2009 23. Prince MR, Zhang H, Morris M, MacGregor JL, Grossman ME,
21. Martin DR, Krishnamoorthy SK, Kalb B, Salman KN, Sharma P, Silberweig J, DeLapaz RL, Lee HJ, Magro CM, Valeri AM: Inci-
Carew JD, Martin PA, Chapman AB, Ray GL, Larsen CP, Pearson dence of nephrogenic systemic fibrosis at two large medical centers.
TC: Decreased incidence of NSF in patients on dialysis after chang- Radiology 248: 807– 816, 2008
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

Syllabus
Clinical Pharmacology for the Nephrologist
Suzanne K. Swan, MD,* Ali Olyaei, PharmD,† and Domenic A. Sica, MD‡
*Division of Nephrology, Hennepin County Medical Center and University of Minnesota,
Minneapolis, Minnesota; †Division of Nephrology and Hypertension, Oregon State University,
College of Pharmacy/Oregon Health & Sciences University, Portland, Oregon; and ‡Division
of Nephrology, Virginia Commonwealth University, Richmond, Virginia

Learning Objectives: route” from the body for myriad pharmacologic agents
1. To understand basic pharmacokinetic principles and toxins, making the kidney especially vulnerable to
injury from such agents, as shown in Figure 1 (2).
that must be considered when prescribing medica-
Thus, understanding mechanisms of nephrotoxicity,
tions for patients with decreased kidney function
whether kidney specific, patient specific, or drug/toxin
2. To understand the effect that dialysis techniques
specific, as well as interventions that either minimize
have on drug elimination
or exacerbate such nephrotoxic injury is critical for the
3. To understand various drug dosing strategies that
prescribing nephrologist. This section reviews basic
may be used for patients who undergo dialysis
pharmacokinetic principles that are altered by renal
therapy impairment, and a step-wise approach to dosage ad-
4. To understand common drug-induced adverse justment is presented. Specific drug-dosing recom-
events that are encountered in patients with chronic mendations for a given pharmaceutical agent are not
kidney disease and renal transplant recipients presented here but can be found in tabular form in a
5. To recognize and understand various drug– drug variety of reference sources listed in Table 1 as well as
interactions that may be encountered in renal package inserts. The most current and comprehensive
transplant recipients review of pharmacologic principles can be found in
The metabolism and elimination of a majority of the November 2009 issue of Clinical Pharmacology &
pharmacologic agents depend on normal renal func- Therapeutics.
tion. In addition, many compounds have pharmacolog-
ically active metabolites that undergo renal excretion.
Somewhat surprising, Zhang et al. (1) recently re- In general, dosage reduction is indicated in
ported a study in which a large percentage of newly patients with CKD when >30% of a drug or
approved drugs that do not undergo renal elimination an active metabolite appears unchanged
from the body nonetheless warranted dosage reduction in the urine.
in patients with renal impairment. As such, dosage
adjustments may be required in patients with renal
insufficiency, both acute and chronic, across a wide Altered Pharmacokinetic Principles
spectrum of disease states and drug categories. Famil- In general, dosage reduction is indicated in pa-
iarity with basic pharmacologic principles is necessary tients with chronic kidney disease (CKD) when ⱖ30%
when approaching dosimetry in renal impairment set- of a drug or an active metabolite appears unchanged in
tings, because patients with kidney disease exist the urine. Metabolites of drugs are particularly prob-
within a narrow therapeutic window in which drug lematic because information is rarely available regard-
accumulation (overdose) as well as subtherapeutic ing their presence and whether they are pharmacolog-
dosing must be avoided. Complicating dosing regi- ically active. Unanticipated responses to drugs should
mens further is that the urinary tract is the major “exit raise suspicions that metabolites may be accumulating
220
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 221

Oral, intramuscular, inhaled, or subcutaneous routes, in


contrast, often result in smaller percentages of a given
dose reaching target tissues. Furosemide, for example,
demonstrates 100% bioavailability when given intrave-
nously but is only on average 50% bioavailable (range
10 to 90%) when given orally. This explains the
common practice of doubling the dose of furosemide
when converting a patient from intravenous to oral
dosing regimens. Drug absorption dictates bioavail-
ability and may be altered in patients with renal
disease. For example, absorption may be reduced by
gastroparesis-induced nausea and vomiting secondary
to diabetic neuropathy or aging. Concomitant admin-
istration of medications such as phosphate-binding
Figure 1. Kidney-specific factors that enhance nephrotoxic agents (all classes) can form insoluble complexes with
risk. First, high renal blood flow (RBF) increases drug various drugs such as quinolone antibiotics, reducing
delivery to the kidney. High metabolic rates of cells increase their absorption and slowing gut motility (3). Gastric
risk for nephrotoxicity with certain drugs. Renal biotrans- acid–lowering agents such as proton pump inhibitors
formation of drugs to toxic metabolites and reactive oxygen
species (ROS) overwhelms local antioxidants. Increased and histamine2 blockers as well as phosphate-binding
concentration of drugs in the medulla and interstitium agents can increase stomach pH, which may impair
causes nephrotoxicity. Finally, apical uptake of certain drug absorption as well (e.g., atazanavir, antidepres-
drugs (aminoglycosides, sucrose) and basolateral transport sants, oral iron).
of drugs through the organic anion transporter (OAT; teno-
fovir) and organic cation transporter (OCT; cisplatin) trans- The volume of distribution (Vd) of a drug does
porters increases renal toxicity. Reprinted from reference 2, not refer to a specific anatomic “compartment” but
with permission. rather all of the compartments in which a given com-
pound may be found: Plasma, water, fat, red blood
or toxic, particularly with newly approved compounds, cells, intracellular versus extracellular binding sites,
for which little clinical experience exists. and tissue binding. In general, plasma concentrations of
Bioavailability and drug absorption, volume of a drug tend to correlate inversely with its Vd. Hydrophilic
distribution, protein binding, metabolism or biotrans- drugs, for example, will demonstrate a lower Vd in
formation, and renal excretion are key pharmacoki- volume-contracted states with a corresponding rise in
netic parameters that are altered by decreased kidney plasma concentrations, whereas edema and ascites tend
function. Bioavailability refers to the fraction (%) of to increase Vd values and lower measured drug levels.
a given dose that reaches the systemic circulation and Clinically, Vd can be used to calculate the dosage re-
is primarily determined by the rate and route of ad- quired to achieve a desired drug level systemically by
ministration of the drug. In general, bioavailability is multiplying the desired concentration (mg/L) by the Vd
considered to be 100% after intravenous administration, (L/kg). For example, a dose of 2.5 mg/kg of the
because all of the dose reaches the systemic circulation. aminoglycoside tobramycin would be administered to

Table 1. Reference sources for dosing adjustments in patients with CKD


Drug Prescribing in Renal Failure: Dosing Guidelines for http://www.acponline.org (PDA version available)
Adults, 5th Ed.
MICROMEDEX Healthcare Series
Up to Date, Inc. http://www.utdol.com/online/index.do
Lexi-Comp, Inc. http://www.lexi.com/individuals/physicians
EPOCRATES http://www.epocrates.com
National Kidney Disease Education Program http://www.nkdep.nih.gov
FDA MedWatch http://www.fda.gov/medwatch
The Medical Letter on Drugs and Therapeutics
222 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

achieve a peak concentration of 10 mg/L given the Vd


of 0.25 L/kg of tobramycin. The Vd for many drugs There is mounting evidence that renal im-
can be found in tabular form in pharmaceutical refer- pairment also affects hepatic drug metabo-
ences or package inserts but generally reflects values lism and disposition with a greater impact
that are obtained from healthy individuals, not patients on nonrenal clearance and bioavailability
with CKD, who often display significant alterations in than was previously recognized.
Vd, as in the case of digoxin. Uremic waste products
are thought to displace digoxin from cellular binding There is mounting evidence that renal impair-
sites, thereby reducing the Vd of digoxin and increas- ment also affects hepatic drug metabolism and dispo-
ing plasma concentrations. sition with a greater impact on nonrenal clearance and
Protein binding can affect the Vd of a drug and bioavailability than was previously recognized. Ani-
the methods by which the concentrations of the drug mal data have shown a significant downregulation of
are measured. Compounds circulate in both protein- hepatic cytochrome P450 metabolism in CKD, and
bound and unbound forms, although it is only the additional evidence suggested the presence of a circu-
unbound, or “free,” fraction that is biologically active. lating inhibitor of hepatic metabolic pathways that
Most drug assays, however, represent the “total” could conceivably be removed by dialysis (5). How
amount of drug present (both bound and unbound). this will ultimately affect drug dosing in patients with
Renal impairment tends to decrease protein binding of CKD remains to be seen, but more compounds that do
drugs because accumulated organic waste products not undergo renal elimination from the body will
displace these compounds from their binding proteins. likely warrant dosage adjustment when renal impair-
This, in turn, results in a larger proportion of the drug ment is present, as recently reported (1).
circulating in its unbound (free) or active form but also
makes more drug available for hepatic metabolism/
Stepwise Approach to Dosimetry
elimination. As such, following free levels rather than The following stepwise approach provides a
total concentrations of a given drug may be prudent, framework for dosage adjustment in patients with
because the latter may be unpredictable given the renal impairment (Figure 2). It must be emphasized
reduced bound fraction (see the Older Anticonvulsant that this approach simply represents a starting point
Drugs section). from which dosimetry must be closely monitored and
Biotransformation or metabolism of a drug modified on a patient-by-patient basis.
refers to its biochemical conversion from one chemical
form to another, usually mediated by hepatic enzy- Step 1: History and Physical Examination
matic pathways such as oxidation, reduction, acetyla- A history and physical examination represent the
tion, glucuronidation, or hydrolysis. Pharmacologi- first step in determining the need for dosage adjust-
cally active metabolites of inactive prodrugs or ments in any patient. Renal impairment should be
metabolites with toxic effects that depend on renal defined as acute or chronic and the cause ascertained
excretion to be eliminated from the body may be when possible to determine reversibility. A history of
formed. For example, many angiotensin-converting drug intolerance, allergy, or nephrotoxicity is key. The
enzyme inhibitors are inactive prodrugs that are con- medication list, both prescribed and nonprescribed,
verted by the liver to active metabolites (e.g., lisinopril should be reviewed for both potential nephrotoxins
3 lisinoprilat), which undergo renal elimination. and drug– drug interactions. The dosage of many drugs
Thus, the half-life of lisinoprilat is markedly pro- depends on a patient’s weight or size but may present
longed in acute renal failure settings and may delay a clinical quandary regarding which weight to use:
“recovery” of glomerular filtration given its site of Ideal body weight (IBW) versus actual body weight or
action. The narcotic meperidine undergoes hepatic an intermediate value. IBW can be calculated using
metabolism to normeperidine, which has no narcotic the patient’s height. For women, IBW is 45.5 kg plus
activity but lowers the seizure threshold and relies on 2.3 kg for every 2.5 cm over 152.0 cm. For men, IBW
the kidney for elimination from the body. As such, is 50.0 kg plus 2.3 kg for every 2.5 cm over 152.0 cm
meperidine should not be used in patients with CKD (6). Body mass index (weight in kg/height in m2) has
given its ability to induce seizure activity (4). been used increasingly as a measure of obesity but is
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 223

(Scr) and blood urea nitrogen values are crude markers


of kidney function at best, attempts to define better
measures of GFR have long been pursued. Histori-
cally, it is important to remember that dosage recom-
mendations for many drugs were generated at a time
7
when only the Cockcroft-Gault (CG) equation (7) was
being used to estimate creatinine clearance (Ccr).
8 CG-derived Ccr ⫽ [(140 ⫺ age) ⫻ (wt in kg)]/
[72 ⫻ Scr] ⫻ 0.85 (if female)
More recently, the Modification of Diet in Renal
Disease (MDRD) study equation (further modified as
the isotope dilution mass spectroscopy–traceable four-
variable equation) (8) has been derived in an effort to
estimate GFR more accurately because Ccr represents
an overestimation:
MDRD-GFR ⫽ 175 ⫻ Scr⫺1.154⫻ age⫺0.203⫻
1.2 (if black) ⫻ 0.74 (if female)
MDRD-derived GFR values, however, have not
been applied to pharmacokinetic studies from which
dosing recommendations often arise. Numerous pub-
lications debating whether the CG or MDRD equation
should be used to estimate kidney function for the
purpose of dosage adjustment have appeared, yet no
conclusions have been reached (9 –11). In fact, the
Figure 2. Stepwise approach to dosimetry. Food and Drug Administration has not finalized its
most recent draft guidance on the conduct of pharma-
generally not used to calculate drug dosage. Of note, cokinetic trials of patients with renal impairment given
drug dosages are generally established in people of the lack of consensus in the literature. A review of this
moderate size (i.e., BMI 18 to 30), not morbidly obese literature is beyond the scope of this issue of Neph-
individuals, so Food and Drug Administration–ap- SAP, but, in general, the MDRD equation underesti-
proved drug dosages may be grossly inadequate in this mates GFR, particularly at advanced stages of renal
population. Alternatively, some medications, particu- impairment (i.e., ⬍30 ml/min) and in normal individ-
larly chemotherapy agents, are based on body surface uals, whereas the CG equation overestimates it. All of
area, which can be calculated by various equations, this is further complicated by newer assays to measure
including the Dubois method (http://www.halls.md/ creatinine (isotope dilution mass spectroscopy–trace-
body-surface-area/refs.htm), which is discussed else- able method versus older methods), which in turn
where in this issue of NephSAP. Volume status is affect any mathematical formula in which the values
important to assess, because extreme conditions can are used. Moving forward, MDRD-derived GFR val-
significantly alter both the Vd and drug concentrations. ues should be tested against CG-derived values in
For example, severe “third-spacing” with 10 or 20 L of pharmacokinetic Phase I studies involving new com-
ascites can increase the Vd of a drug and thereby lower pounds to provide greater clarification. Of note, in
serum concentrations, whereas extensive burns can
patients with acute renal failure (particularly oliguric
have the opposite effect (i.e., marked decrease in Vd
or anuric), the prescribing physician should assume a
with subsequent elevation in drug levels).
GFR of ⬍10 ml/min for the purposes of drug dosage
Step 2: Assess Renal Function adjustment. Alternatives to measure kidney function
Drug elimination by the kidney correlates with involving “plasma disappearance” techniques such as
GFR; therefore, it is logical to use GFR to gauge iohexol- or iothalamate-derived methods exist but may
adjustments in drug dosage. Because serum creatinine not be clinically feasible for many prescribers (12).
224 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

Step 3: Determine Loading Dose give a modified dose at a modified interval. Thus, one
A loading dose of a drug is given to achieve of the following dosing regimens would be possible
steady-state conditions more rapidly, because it gen- for a patient with a 50% reduction in kidney function
erally requires three to four half-lives of a given drug when administering a drug with a standard dose of 300
to achieve steady-state concentrations. Attaining steady- mg every 6 hours daily:
state levels could be significantly prolonged in renal
Y 150 mg every 6 hours (varying dose)
impairment as a result of prolongation of the half-life Y 300 mg every 12 hours (varying interval)
of a drug. In general, the standard loading dose that is Y 200 mg every 8 hours (combination)
given to patients with normal renal function should be
Which method is used depends on the underlying
given to patients with renal impairment to reach ther-
disease state being treated and the therapeutic window
apeutic drug levels rapidly. A loading dose can be
of the drug. For example, the varying dose method
calculated (discussed already with tobramycin exam-
might be preferable when using a drug with a narrow
ple) by the following formula (Cp is the desired
therapeutic concentration range (anticonvulsant or anti-
plasma concentration):
arrhythmic agents) in which more constant levels are
Loading dose ⫽ Vd (L/kg) ⫻ Cp (mg/L) ⫻ IBW (kg)
desired and subtherapeutic periods are avoided. Con-
Digoxin dosing represents one exception to this
versely, the varying interval method may be preferred
rule. The Vd for digoxin is reduced by 50 to 75% in
when dosing aminoglycoside antibiotics, which have a
severe kidney impairment that is great enough in mag-
long postantibiotic effect whereby long periods of low
nitude to warrant a corresponding reduction in loading
drug levels are not problematic and bacterial killing
dose. Thus, given the direct proportionality between
depends on high peak concentrations (15) (more detailed
loading dose and Vd, one could easily calculate a
discussion can be found in infectious disease textbooks).
“modified” loading dose by the following:
Modified loading dose ⫽ [patient’s Vd/normal Step 5: Drug-Level Monitoring
Vd] ⫻ standard loading dose Varying the dosage or dosing interval for a given
One area of controversy involves whether to use therapeutic agent may not be sufficient to avoid tox-
actual body weight versus IBW or something midway icity and guarantee therapeutic efficacy in patients
between these two values when calculating loading with renal failure. Monitoring drug levels can greatly
doses. Vancomycin dosing, for example, is one that enhance patient care, particularly in the setting of renal
should be based on actual body weight, not IBW, as impairment. Interpreting drug concentrations requires
recently reviewed (13,14). knowledge of the exact dosage given, the route of
administration, whether the Vd or protein binding of
Step 4: Determine Maintenance Dose the drug is altered, the elapsed time from the last dose,
The maintenance dose is the dosing regimen that and the elimination half-life of the drug. Drug levels
maintains steady-state concentrations and can be may be monitored after an appropriate loading dose or
achieved by one of three methods: Varying dose three to four maintenance doses have been given to
method, varying interval method, or a combination of ensure that steady-state concentrations have been
the two. In the varying dose approach, a drug is reached. As a rough rule of thumb, peak drug levels
administered at the same interval used in patients with reflect the highest drug concentration achieved after an
normal renal function, but the actual dose is reduced in initial distribution phase and tend to correlate with
proportion to the patient’s degree of renal impairment efficacy. Trough levels are generally obtained imme-
as follows (assume normal Ccr of 100 ml/min): diately before the next dose to determine the lowest
Modified maintenance dose ⫽ [patient’s Ccr/ concentration in the blood, thus reflecting systemic
normal Ccr] ⫻ standard maintenance dose clearance, and generally reflect toxicity/accumulation.
Conversely, the dosing interval can be length- Drug monitoring must include ongoing clinical assess-
ened (modified dosing interval) while keeping the ment of the patient because toxicity can occur even
actual dose the same as the standard dose: when levels are within the “therapeutic” range or
Modified dosing interval ⫽ [normal Ccr/pa- undetectable. For example, digoxin toxicity can occur
tient’s Ccr] ⫻ standard dosing interval in the setting of therapeutic levels when hypokalemia
Or one can combine these two approaches and or metabolic alkalosis is present. It is important to
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 225

remember that pharmacokinetic half-life (i.e., disap-


pearance from blood) does not necessarily equal “ef-
fect” half-life, as is the case with clopidogrel, a platelet
aggregation inhibitor. Clopidogrel has an elimination
half-life that is measured in hours, whereas its effect
half-life (inhibition of platelet aggregation) extends to
days and even weeks. The assumption that platelet
function has been restored because drug can no longer
be measured in the blood could prove lethal for a
Figure 3. Compounds may traverse numerous compart-
patient who undergoes a surgical procedure if, in fact, ments in the body before elimination.
the antiplatelet effect is still present. Similarly, for
some drugs such as aminoglycoside antibiotics, drug-
level monitoring has not prevented toxicity because of ably after dialysis) and/or supplemental dosing if an
the poor correlation between serum levels and uptake agent is substantially cleared by dialysis (⬎30 to 40%
into tissues such as the proximal tubular epithelial cell increased extracorporeal clearance). The dialyzability
(nonoliguric acute renal failure) and inner ear (vestib- of a drug, often unknown, can be approximated by
ular ototoxicity). Last, the pharmacodynamic effects considering three factors: The drug, the dialysis mem-
of a drug may be amplified in patients with CKD or brane, and dialysis method used (Table 2). First, phar-
ESRD as a result of increased sensitivity at the target macologic characteristics of a drug—size, water solu-
organ (e.g., excessive sedation and respiratory sup- bility, protein binding, Vd, charge, and renal versus
pression with narcotics). nonrenal clearance— determine the extent to which it
may be removed. Thus, small (⬍500 Da), water-
Drug Clearance in Dialysis soluble (appear unchanged in urine), unbound drugs
The following information regarding dialytic
with small distribution volumes would likely be re-
clearance of drugs is detailed in the recent issue of
moved from the body by dialysis. An example of this
Clinical Pharmacology & Therapeutics (referred to in
is aminoglycoside antibiotics. Conversely, drugs that
the introduction).
exhibit a large Vd (often highly protein or tissue
Patients who undergo dialysis therapy, both in-
bound) are not readily dialyzed, because only a small
termittent and continuous, require special attention
portion of the total amount of drug in the body circu-
with regard to scheduling drug administration (prefer-
lates in the vascular space with a much larger portion
“out of reach” of dialytic removal. One can remove all
Table 2. Factors that affect drug dialyzability of the drug in the vascular space with a dialysis
treatment, but it is rapidly replenished with the large
Drug amount of drug found in various tissue compartments,
size (molecular weight)
as illustrated in Figure 3. Cyclosporine, digoxin, and
% protein bound
volume of distribution tricyclic antidepressants are examples of drugs that
water solubility (% of dose appearing “unchanged” in exhibit such pharmacologic qualities.
urine) Second, a dialysis membrane with a larger sur-
Membrane face area and greater permeability (larger pore size)
size (surface area) would be expected to remove more drug, larger com-
permeability (pore size) pounds (up to 40,000 Da in some cases), and some
composition (polysulfone, cellulose-based, etc.) protein-bound agents. Such membranes are referred to
drug-membrane–binding characteristics (if any)
as high-flux dialysis membranes and include polysul-
Dialysis method
dialysis (drug removal by diffusion) fone, polyacrylonitrile, and cellulose triacetate. This is
hemofiltration (drug removal by convection) pertinent because vancomycin, for example, is signif-
duration of procedure icantly removed by such dialysis membranes in con-
blood and dialysate flow rate trast to older, low-flux membranes (cellulose). As
ultrafiltration rate such, vancomycin dosing in patients who undergo high-
replacement solution location (pre- or postfilter) flux dialysis has become more complicated, mandating
226 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

higher dosages, more frequent dosing, and more drug- and the pharmacologic principles that apply. Nonethe-
level monitoring. A review by Pallotta and Manley (14) less, a paucity of information exists regarding altered
in addition to many previous reports detailed the phar- pharmacology in the critical care setting, where acute
macokinetic issues involved in vancomycin dosing for kidney failure, hepatic insufficiency, altered volume
patients who undergo dialytic therapy (16,17). status, hypoalbuminemia, multiple drug interactions,
Last, assuming that a particular drug can be and the use of various dialysis techniques further
removed by dialysis, the methods used during dialysis, complicate drug-dosing regimens. This realization fur-
such as blood flow rate, dialysate flow rate, ultrafil- ther emphasizes the need to individualize each pa-
tration rate, and duration of treatment, directly corre- tient’s therapy by continuous clinical assessment
late with the amount of drug removed. With the advent whereby “more” antimicrobial therapy rather than
of short-daily and nocturnal hemodialysis, dosing reg- “less” may be prudent given the danger of ineffective
imens may require further adjustment given the overall therapy in the intensive care unit setting (21,22).
increased clearance achieved by these methods (18).
In the case of continuous renal replacement therapy References
(CRRT), combining hemofiltration with dialysis will 1. Zhang Y, Zhang L, Abraham S, Apparaju S, Wu TC, Strong JM,
Xiao S, Atkinson AJ Jr, Thummel KE, Leeder JS, Lee C, Burckart
remove pharmaceutical agents by convection (hemofil-
GJ, Lesko LJ, Huang SM: Assessment of the impact of renal
tration) and diffusion (dialysis), resulting in dosing strat- impairment on systemic exposure of new molecular entities: Eval-
egies that fall somewhere between those that are used in uation of recent new drug applications. Clin Pharmacol Ther 85:
outpatient intermittent hemodialysis settings (GFR ⬍10 305–311, 2009
2. Perazella MA: Renal vulnerability to drug toxicity. Clin J Am Soc
ml/min) and those that are used in mild impairment (GFR Nephrol 4: 1275–1283, 2009
50 to 80 ml/min). Convection removes unbound drug in 3. Lomaestro BM, Bailie GR: Quinolone-cation interactions: A re-
view. DICP 25: 1249 –1258, 1991
the serum, and this amount can be calculated as follows: 4. Hassan H, Bastani B, Gellens M: Successful treatment of norme-
Amount removed ⫽ serum concentration ⫻ unbound peridine neurotoxicity by hemodialysis. Am J Kidney Dis 35: 146 –
fraction ⫻ ultrafiltration rate ⫻ treatment duration 149, 2000
5. Dreisbach AW, Lertora JJ: The effect of chronic renal failure on
The amount of drug removed by diffusion during hepatic drug metabolism and drug disposition. Semin Dial 16:
concurrent dialysis can be added to this amount to 45–50, 2003
determine dosage regimens during CRRT. The loca- 6. Brier ME, Aronoff GR, eds.: Drug Prescribing in Renal Failure:
Dosing Guidelines for Adults, 5th Ed., Philadelphia, ACP-ASIM,
tion of replacement solutions (pre- versus postfilter) in 2007
the system can also affect drug clearance. Prefilter 7. Cockcroft DW, Gault MH: Prediction of creatinine clearance from
replacement fluids may result in higher ultrafiltration serum creatinine. Nephron 16: 31– 41, 1976
8. Levey AS, Coresh J, Greene T, Stevens LA, Zhang YL, Hendriksen
rates and higher midsize solute clearance, whereas S, Kusek JW, Van Lente F, Chronic Kidney Disease Epidemiology
postfilter replacement fluid can increase filtration frac- Collaboration: Using standardized serum creatinine values in the
tion. A recent review by Susla (19) discussed the modification of diet in renal disease study equation for estimating
glomerular filtration rate. Ann Intern Med 145: 247–254, 2006
impact of CRRT on drug removal. 9. Stevens LA, Nolin TD, Richardson MM, Feldman HI, Lewis JB,
Rodby R, Townsend R, Okparavero A, Zhang YL, Schmid CH,
Levey AS: Comparison of drug dosing recommendations based on
measured GFR and kidney function estimating equations. Am J
A paucity of information exists regarding Kidney Dis 54: 33– 42, 2009
altered pharmacology in the critical care 10. Moranville MP, Jennings HR: Implications of using Modification of
Diet in Renal Disease versus Cockcroft-Gault equations for renal
setting, where acute kidney failure, he-
dosing adjustments. Am J Health Syst Pharm 66: 154 –161, 2009
patic insufficiency, altered volume sta- 11. Golik MV, Lawrence KR: Comparison of dosing recommendations
tus, hypoalbuminemia, multiple drug in- for antimicrobial drugs based on two methods for assessing kidney
function: Cockcroft-Gault and Modification of Diet in Renal Disease.
teractions, and the use of various dialysis Pharmacotherapy 28: 1125–1132, 2008
techniques further complicate drug-dos- 12. Gaspari F, Perico N, Ruggenenti P, Mosconi L, Amuchastegui CS,
ing regimens. Guerini E, Daina E, Remuzzi G: Plasma clearance of nonradioactive
iohexol as a measure of glomerular filtration rate. J Am Soc Nephrol
6: 257–263, 1995
13. Pai MP, Bearden DT: Antimicrobial dosing considerations in obese
adult patients. Pharmacotherapy 27: 1081–1091, 2007
14. Pallotta KE, Manley HJ: Vancomycin use in patients requiring
Pea et al. (20) published an exhaustive review of hemodialysis: A literature review. Semin Dial 21: 63–70, 2008
antimicrobial therapy for patients who undergo CRRT 15. Nicolau DP, Freeman CD, Belliveau PP, Nightingale CH, Ross JW,
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 227

Quintiliani R: Experience with a once-daily aminoglycoside program tween a ␤-lactam antibiotic and its lactamase inhibitor
administered to 2,184 adult patients. Antimicrob Agents Chemother
39: 650 – 655, 1995
may affect their use in renal failure settings. In the
16. Rybak MJ: The pharmacokinetic and pharmacodynamic properties of case of ampicillin/sulbactam, both components are
vancomycin. Clin Infect Dis 42: S35–S39, 2006 eliminated by the kidney and have similar half-lives
17. Pai AB, Pai MP: Vancomycin dosing in high flux hemodialysis: A
limited-sampling algorithm. Am J Health Syst Pharm 61: 1812–1816,
and volumes of distribution across all levels of renal
2004 function and during dialysis. This allows for an equiv-
18. Decker BS, Mueller BA, Sowinski KM: Drug dosing considerations alent dosage reduction (or dosing interval extension)
in alternative hemodialysis. Adv Chronic Kidney Dis 14: e17– e26,
2007 for each component, thereby maintaining lactamase
19. Susla GM: The impact of continuous renal replacement therapy on inhibition during dosing intervals. The same is true for
drug therapy. Clin Pharmacol Ther 86: 562–565, 2009 piperacillin/tazobactam. In contrast, ticarcillin/clavu-
20. Pea F, Viale P, Pavan F, Furlanut M: Pharmacokinetic considerations
for antimicrobial therapy in patients receiving renal replacement lanate may have greater pharmacokinetic disparity
therapy. Clin Pharmacokinet 46: 997–1038, 2007 between the two components such that clavulanate
21. Bouman CS: Antimicrobial dosing strategies in critically ill patients levels may be insufficient to protect ticarcillin from
with acute kidney injury and high-dose continuous veno-venous
hemofiltration. Curr Opin Crit Care 14: 654 – 659, 2008 lactamase-producing organisms when the dosing inter-
22. Schetz M: Drug dosing in continuous renal replacement therapy: val is extended in patients with ESRD (1). In general,
General rules. Curr Opin Crit Care 13: 645– 651, 2007
excess levels of penicillins may be associated with
central nervous system toxicity, including seizures,
Antimicrobial Agents coma, and myoclonus. Similarly, the carbapenem imi-
Antibacterial Agents penem can lower the seizure threshold with accumu-
Much of what follows in this brief overview lation when dose reduction is not performed for pa-
comes from multiple references in older literature; tients with renal impairment (2).
pharmacologic properties of specific agents can be Doxycycline is one tetracycline that does not
reviewed by the reader on a case-by-case basis. Im- seem to have an antianabolic effect previously re-
portant concepts that are needed by the prescribing ported for older agents in this category and thus is safe
physician to design a “pharmacologic approach to to use in patients with CKD. Nitrofurantoin, com-
patients with kidney impairment” follow. monly prescribed for chronic bacterial suppression
therapy in urologic disorders, should not be used in
CKD to avoid peripheral neuropathy attributed to toxic
Many antibacterial agents appear in the metabolite accumulation (3).
urine unchanged (water soluble), are small Aminoglycoside antibiotics (gentamicin, tobra-
molecules (<500 Da), and are not highly mycin, netilmicin, amikacin, and streptomycin) are
protein bound, thereby necessitating dos- less commonly used because of nephrotoxicity and
age reduction in patients with CKD as well ototoxicity but are nonetheless effective bactericidal
as supplementation after dialysis. agents and relatively inexpensive. The kidney freely
filters aminoglycosides and then resorbs filtered drug
by megalin-mediated endocytosis in the proximal tu-
bule. High intracellular concentrations lead to lysoso-
Many antibacterial agents appear in the urine mal, Golgi apparatus, and endoplasmic reticulum dis-
unchanged (water soluble), are small molecules (⬍500 ruption in these tubular epithelial cells. Resulting cell
Da), and are not highly protein bound, thereby neces- death of proximal tubular epithelia leads to nonoligu-
sitating dosage reduction in patients with chronic kid- ric renal failure. Vestibular and cochlear abnormalities
ney disease (CKD) as well as supplementation after are linked to injury of hair cells within the inner ear
dialysis. ␤-Lactam agents, such as penicillins, cepha- bathed by aminoglycoside-containing endolymph.
losporins, carbapenems, and monobactams, are exam- Once-daily dosing has been shown to minimize neph-
ples of such compounds. Combining certain penicil- rotoxicity (especially when GFR is ⬍60 ml/min) and
lins with ␤-lactamase inhibitors such as sulbactam, takes advantage of high peak levels delivering maxi-
tazobactam, and clavulanate may minimize develop- mal bactericidal effect, which continues during longer
ment of antibiotic resistance and extend their spectrum dosing intervals as a result of the postantibiotic effect
of activity. Different pharmacokinetic properties be- displayed by these agents (4). Because nephrotoxicity
228 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

correlates with frequency of dosing and high trough some of these clinical abnormalities that are schemat-
levels, once-daily or once-every-other-day dosing for ically illustrated in the review.
patients with CKD is desirable. It is important to
remember that implanted aminoglycoside “reservoirs,” Antifungal Agents
which are used in orthopedic procedures, can also Antifungal agents include the azoles (flucon-
lead to acute toxicity (5). azole, itraconazole, voriconazole, and posaconazole),
Allergic interstitial nephritis has classically been the echinocandins (caspofungin, micafungin, and anidu-
associated with exposure to penicillins, cephalospo- lafungin), and the amphotericin family of polyenes
rins, and sulfa-containing agents. Fluoroquinolone an- (amphotericin B, amphotericin B-liposome, amphoter-
tibiotics such as ciprofloxacin, levofloxacin, and oth- icin B–lipid complex, and amphotericin B– cholesteryl
ers should be added to this list given numerous case sulfate complex). An extensive review of antifungal
reports of quinolone-induced allergic interstitial ne- agents in the treatment of invasive fungal infections
phritis (6). Vancomycin dosing is discussed in the has appeared recently (8). Table 4 is not an exhaustive
previous Stepwise Approach to Dosimetry and Drug list of all antifungal agents and does not include Food
Clearance in Dialysis sections. and Drug Administration–approved dosage guidelines
Recently, Zietse et al. (7) reviewed fluid, elec- but simply highlights clinical issues that pertain to
trolyte, and acid-base disorders that are associated patients with CKD. Detailed information can be found
with antibiotic therapy. Their work is a welcome online from the Infectious Diseases Society of Amer-
addition to the renal pharmacology literature with its ica (http://www.idsociety.org) as well as previously
emphasis on various receptors, transporters, channels, mentioned references (Table 1).
and/or pores that are found in renal tubular epithelial Amphotericin B
cell membranes and can be affected by various anti- All antifungal agents disrupt the fungal cell
microbials. Thus, renal tubular function can be af- membrane by either binding to ergosterol in the lipid
fected by antimicrobials often without a concurrent bilayer (amphotericin-based agents) or inhibiting its
rise in serum creatinine or decline in GFR. These synthesis. This leads to an inability to maintain cell
so-called “tubulopathies” that are associated with var- membrane integrity and subsequent leakage of cell
ious antimicrobials include sodium wasting, potassium contents. Unfortunately, much of the toxicity that is
wasting, renal tubular acidosis, and Fanconi syndrome associated with amphotericin therapy is linked to this
as well as nephrogenic diabetes insipidus. Table 3 lists mechanism of action, because these compounds can

Table 3. Tubulopathy syndromes induced by antimicrobials (7)


Syndrome Description
Aminoglycosides and tetracyclines
proximal RTA or Fanconi syndrome Normal anion gap metabolic acidosis; hypokalemia; uric acid, glucose,
amino acid, phospho-wasting
Bartter-like syndrome Metabolic alkalosis with K⫹, Ca⫹, Mg⫹, Na⫹ wasting
Amphotericin B compounds
distal RTA (H⫹ “backleak”) Normal anion gap metabolic acidosis
hypokalemia K⫹ wasting; distal RTA
polyuria/hypernatremia Nephrogenic diabetes insipidus
Trimethoprim
distal RTA Normal anion gap metabolic acidosis
blockade of ENaC Hyperkalemia
Penicillins
hypokalemia High urine K⫹, low urine Cl⫺
hyperkalemia High K⫹ content in setting of poor kaliuresis
Linezolid and tetracyclines
lactic acidosis High anion gap metabolic acidosis
ENaC, epithelial sodium channels; RTA, renal tubular acidosis.
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 229

Table 4. Antifungal agents


Agent Dosage Reduction Adverse Effects
Echinocandins
caspofungin No
micafungin No
anidulafungin No
Amphotericin B
amphotericin B lipid complex No Lipid formulations less nephrotoxic
than amphotericin B, but tubular
cell injury still present and
decreased GFR; RTA; NDI; K⫹
and Mg2⫹ wasting. Infusion
reaction specific for lipid
preparations
amphotericin B liposomal No
amphotericin B No
cholesterylsulfate
Azoles
fluconazole Yes (Ccr ⬍50 ml/min); supplement after HD Alopecia
posaconazole No
itraconazole (cyclodextrin) Avoid intravenous form if Ccr ⬍30 ml/min; Hyperaldosteronism
no change if oral
voriconazole (cyclodextrin) Avoid intravenous form if Ccr ⬍50 ml/min; CNS (cyclodextrin versus drug),
no change if oral visual hallucinations, agitation,
myoclonus; photopsia (?
cyclodextrin); colored light,
flashing light; increased
creatinine
CNS, central nervous system; NDI, nephrogenic diabetes insipidus; RTA, renal tubular acidosis.

bind to cholesterol moieties in human cell membranes significant cost (9). A unique infusion reaction has
as well. Renal tubular epithelial cell function is thus been observed with liposomal amphotericin B and
impaired, leading to potassium and magnesium wast- consists of chest pain, dyspnea, hypoxia, abdominal
ing, distal renal tubular acidosis from H⫹ backleak, pain, and urticaria. Amphotericin is highly protein
and concentrating defects with nephrogenic diabetes bound, has a large Vd, and undergoes little renal
insipidus (see Table 3). These tubular defects can elimination from the body. Dialysis therapies have
persist long after antifungal therapy is discontinued. little effect on drug removal in any formulation.
Amphotericin is also thought to cause vasoconstric- Azoles
tion, reducing renal blood flow and contributing to Azoles inhibit synthesis of ergosterol, an essen-
azotemia. This can be ameliorated with volume expan- tial part of the fungal cell membrane. The “tri”-azoles
sion using intravenous sodium chloride before dosing. include fluconazole, itraconazole, voriconazole, and
In addition to nephrotoxicity, infusion-related adverse posaconazole. They have largely supplanted the older
reactions include fever, chills, nausea, vomiting, hy- imidazole ketoconazole because of greater safety, ef-
potension, and tachypnea. The amphotericin class of ficacy, and improved pharmacokinetics. Of note, the
antifungal agents has expanded in the past 10 to 15 solubilizing agent used with voriconazole and itracon-
years with the advent of lipid-based formulations in- azole, cyclodextrin, is eliminated by the kidney, accu-
cluding liposomal amphotericin B, lipid complex, and mulates when GFR is ⬍50 ml/min leading to toxicity,
amphotericin B cholesteryl sulfate in an attempt to and therefore is not recommended for patients with
minimize toxicity. Although the incidence and sever- significant renal impairment. Toxicity can take one of
ity of nephrotoxicity and infusion reactions may be two primary forms: Photopsia (flashing lights, colored
reduced by lipid formulations, both persist and at light) and central nervous system neurotoxicity, in-
230 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

cluding visual and auditory hallucinations, myoclonus, thrombocytopenic purpura are particularly problem-
and agitation (10). Elevation of serum creatinine and atic for patients with CKD, who are often anemic at
photophobia/sun-induced skin reactions have also baseline. Pegylated IFN alfa-2a and -2b can be used
been reported with voriconazole. Intravenous itracon- alone for patients who have CKD with Ccr values ⬍50
azole is no longer available in the United States; ml/min but require close monitoring. For hepatitis B
although cyclodextrin is found in the oral itraconazole infection, lamivudine, telbivudine, and entecavir can
formulation, it does not accumulate in renal impair- also be used but require dosage reduction in patients
ment. with Ccr ⬍50 ml/min (Table 5). Lamivudine (12) does
Echinocandins not seem to be removed by hemodialysis, whereas
A new class of antifungal agents, echinocandins, entecavir (13) and telbivudine (14) should be admin-
inhibits synthesis of ␤-D-glucan, a component of the istered after dialysis (Table 5).
fungal cell wall. Toxicity is much less with these Current opinion on hepatitis B vaccination for
compounds because of the absence of glucan synthesis patients with ESRD varies given the reduced response
enzymes in mammalian cells. Caspofungin, micafun- of such patients to vaccination, which may result from
gin, and anidulafungin are effective against Candida uremia-induced alterations in T lymphocyte and anti-
species, including those that are resistant to the azoles gen-presenting cell behavior (15). Further discussion
as well as Aspergillus. They can be given once daily, of hepatitis B vaccination regimens is beyond the
intravenously, and do not require dosage adjustment in scope of this issue of NephSAP.
renal failure (8). Anti-Influenzal Agents
Antiviral drugs are an important adjunct to influ-
Antiviral Agents enzal vaccinations for treatment and prophylaxis of
A limited number of antiviral therapies for hep- both pandemic and seasonal influenza. Table 6 is not
atitis B, hepatitis C, and influenza are discussed in this an exhaustive list of anti-influenzal agents but pro-
section because of a higher prevalence of such viral vides some guidance with regard to dosing for patients
infections in patients with ESRD. Dosage adjustment with CKD. Amantadine, oseltamivir, and peramivir
of antiretroviral agents and their renal syndromes are undergo renal elimination and warrant dosage reduc-
discussed primarily in tabular form. Other antiviral tion in patients with Ccr ⬍50 ml/min (16).
compounds are discussed in the Immunosuppressive Antiretroviral Agents
Therapies section. The introduction of highly active antiretroviral
Hepatitis B and C Antiviral Agents therapy in the mid-1990s dramatically altered the
IFN alfa-2a and -2b as well as their pegylated course of HIV infections, with marked improvement
formulations can be used for both hepatitis B and C in survival rates. As such, tens of millions of people
infections. Pegylation refers to the attachment of a worldwide now live with HIV infection chronically,
polyethylene glycol side chain to the IFN molecule and this number will continue to grow. Although viral
that reduces the rate of renal elimination, thereby suppression contributes to improved morbidity and
allowing for once-weekly dosing. Guidelines issued mortality rates, long-term exposure to antiretroviral
by the American Association for the Study of Liver
Diseases for the diagnosis, management, and treatment
Table 5. Hepatitis B and C antiviral agents
of hepatitis C was updated in 2009 and can be ac-
cessed through the American Association for the Agent Dosage Reduction Indicated
Study of Liver Diseases web site at http://www.aasld. Hepatitis C
org/practiceguidelines. Currently, the treatment of pegylated IFN alfa-2a and Ccr ⬍50 ml/min and ESRD
-2b
choice for chronic hepatitis C infection is a combina- ribavirin (⫾ pegylated Avoid in Ccr ⬍50 ml/min
tion of pegylated IFN alfa plus ribavirin, both of which IFN)
undergo renal elimination (11). In patients with creat- Hepatitis B
pegylated IFN alfa-2a and Ccr ⬍50 ml/min and ESRD
inine clearance (Ccr) ⬍50 ml/min, ribavirin and the -2b
combination of ribavirin plus IFN alfa agents should lamivudine Ccr ⬍50 ml/min and ESRD
be avoided. Ribavirin has a long list of adverse effects, entecavir Ccr ⬍50 ml/min and ESRD
but hemolytic anemia, red cell aplasia, and thrombotic telbivudine Ccr ⬍50 ml/min and ESRD
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 231

Table 6. Anti-influenzal agents


Agent Treatment Prophylaxis
Oseltamivir Reduce dosage for Ccr ⬍30 ml/min (every day from Reduce for Ccr ⬍30 ml/min (every other day from
twice daily) every day)
Peramivir Dosage reduction Dosage reduction
Amantadine Reduce dosage for Ccr ⬍50 ml/min (every day from Dosage reduce for Ccr ⬍50 ml/min (every day
twice daily) from twice daily)
Zanamivir No change No change

agents poses nephrotoxic injury, and certain agents patients with chronic renal failure: Effects of differential pharmaco-
kinetics on serum bactericidal activity. Pharmacotherapy 14: 147–
must be dosage-adjusted in the setting of CKD. It is 152, 1994
beyond the scope of this syllabus to provide a detailed 2. Seto AH, Song JC, Guest SS: Ertapenem-associated seizures in a
discussion of antiretroviral therapy, but two recent peritoneal dialysis patient. Ann Pharmacother 39: 352–356, 2005
3. White WT, Harrison L, Dumas S 2nd: Nitrofurantoin unmasking
reviews by Kalayjian (17) (drug-dosing recommenda- peripheral neuropathy in a type 2 diabetic patient. Arch Intern Med
tions) and Jao and Wyatt (18) (nephrotoxicity) were 144: 821, 1984
published in January 2010 in Advances in Chronic 4. Nicolau DP, Freeman CD, Belliveau PP, Nightingale CH, Ross JW,
Quintiliani R: Experience with a once-daily aminoglycoside program
Kidney Disease, and the tabular information presented administered to 2,184 adult patients. Antimicrob Agents Chemother
in Tables 7 and 8 has been reprinted with permission 39: 650 – 655, 1995
from these references. 5. Wu IM, Marin EP, Kashgarian M, Brewster UC: A case of an acute
kidney injury secondary to an implanted aminoglycoside. Kidney Int
In general, dosage reductions for kidney im-
75: 1109 –1112, 2009
pairment are not necessary for the following anti- 6. Lomaestro BM: Fluoroquinolone-induced renal failure. Drug Saf 22:
retroviral drug categories/agents: Integrase inhibi- 479 – 485, 2000
7. Zietse R, Zoutendijk R, Hoorn EJ: Fluid, electrolyte and acid-base
tors (raltegravir), fusion inhibitors (enfuvirtide),
disorders associated with antibiotic therapy. Nat Rev Nephrol 5:
CCR5 antagonists (maraviroc), protease inhibitors 193–202, 2009
(atazanivir, darunavir, fosamprenavir, indinavir, 8. Segal BH: Aspergillosis. N Engl J Med 360: 1870 –1884, 2009
lopinavir, nelfinavir, ritonavir, saquinavir, and ti- 9. Caillot D, Reny G, Solary E, Casasnovas O, Chavanet P, Bonnotte B,
Perello L, Dumas M, Entezam F, Guy H: A controlled trial of
pranavir), and nonnucleoside reverse transcriptase in- tolerance of amphotericin B infused in dextrose or in intralipid in
hibitors (delavirdine, efavirenz, etravirine, and nevi- patients with hematologic malignancies. J Antimicrob Chemother 33:
rapine). In the case of nucleoside and nucleotide 603– 613, 1994
10. Pea F, Viale P: Hallucinations during voriconazole therapy: Who is at
reverse transcriptase inhibitors (including zidovudine, higher risk and could benefit from therapeutic drug monitoring? Ther
lamivudine, emtricitabine, stavudine, didanosine, and Drug Monit 31: 135–136, 2009
tenofovir), however, dosage adjustment is warranted 11. Ghany MG, Strader DB, Thomas DL, Seeff LB: Diagnosis, manage-
ment, and treatment of hepatitis C: An update. Hepatology 49:
for patients with CKD. Specifically, nucleotide reverse 1335–1374, 2009
transcriptase inhibitors undergo glomerular filtration 12. Johnson MA, Verpooten GA, Daniel MJ, Plumb R, Moss J, Van
and tubular secretion by proximal tubular epithelial Caesbroeck D, De Broe ME: Single dose pharmacokinetics of lami-
vudine in subjects with impaired renal function and the effect of
cells. Similar to aminoglycoside nephrotoxicity, high haemodialysis. Br J Clin Pharmacol 46: 21–27, 1998
intracellular concentrations of these agents can occur 13. Lok AS, McMahon BJ: Chronic hepatitis B. Hepatology 45: 507–
in the proximal tubular epithelia, resulting in Fanconi 539, 2007
14. Zhou XJ, Myers M, Chao G, Dubuc G, Brown N: Clinical pharma-
syndrome and other tubulopathies. Although protease cokinetics of telbivudine a potent antiviral for hepatitis B, in subjects
inhibitors do not require dosage adjustment, crystallu- with impaired hepatic or renal function. J Hepatol 40: S134, 2004
ria is common and can lead to urolithiasis. More 15. Litjens NH, Huisman M, van den Dorpel M, Betjes MG: Impaired
immune responses and antigen-specific memory CD4⫹ T cells in
detailed dosing information with respect to both he- hemodialysis patients. J Am Soc Nephrol 19: 1483–1490, 2008
patic and renal impairment can be found at http:// 16. Khazeni N, Bravata DM, Holty JE, Uyeki TM, Stave CD, Gould MK:
www.aidsinfo.nih.gov/ContentFiles/AdultandAdoles- Systematic review: Safety and efficacy of extended-duration antiviral
chemoprophylaxis against pandemic and seasonal influenza. Ann
centGL.pdf (19). Intern Med 151: 464 – 473, 2009
17. Kalayjian RC: The treatment of HIV-associated nephropathy. Adv
References Chronic Kidney Dis 17: 59 –71, 2010
1. Hardin TC, Butler SC, Ross S, Wakeford JH, Jorgensen JH: Com- 18. Jao J, Wyatt CM: Antiretroviral medications: Adverse effects on
parison of ampicillin-sulbactam and ticarcillin-clavulinic acid in the kidney. Adv Chronic Kidney Dis 17: 72– 82, 2010
232 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

Table 7. Antiretroviral dosing guidelines with renal insufficiency


Antiretroviral Agent Daily Dosage CrCl (ml/min) Dosage
Abacavir (Ziagen) 300 mg twice daily No dosage adjustment
necessary
Didanosine (Videx EC) ⬎60 kg: 400 mg/d 30 to 59 200 mg
10 to 29 125 mg
CAPD or HD 125 mg
⬍60 kg: 250 mg/d 30 to 59 125 mg
10 to 29 125 mg
⬍10 Not recommended
CAPD or HD Not recommended
Didanosine oral ⬎60 kg: 200 mg twice daily 30 to 59 200 mg
solution (Videx) or 400 mg/d
10 to 29 150 mg
⬍10 100 mg
CAPD or HD 100 mg
⬍60 kg: 250 mg twice daily 30 to 59 150 mg
or 125 mg/d
10 to 29 100 mg
⬍10 75 mg
CAPD or HD 75 mg
Emtricidabine (Emtriva) 200-mg oral capsule daily 30 to 49 200 mg every 48 hours
15 to 29 200 mg every 72 hours
⬍15 200 mg every 96 hours
CAPD or HDa 200 mg every 96 hours
240 mg (24 ml) oral 30 to 49 120 mg every 24 hours
solution daily
15 to 29 80 mg every 24 hours
⬍15 60 mg every 24 hours
CAPD or HDa 60 mg every 24 hours
Lamivudine (Epivir) 300 mg/d or 150 mg twice 30 to 49 150 mg every 24 hours
daily
15 to 29 150 mg ⫻1 then 100 mg every 24 hours
5 to 14 150 mg ⫻1 then 50 mg every 24 hours
⬍5 50 mg ⫻1 then 25 mg every 24 hours
CAPD or HDa 50 mg ⫻1 then 25 mg every 24 hours
Stavudine (Zerit) ⬎60 kg: 40 mg twice daily 26 to 50 20 mg every 12 hours
10 to 25 20 mg every 24 hours
CAPD or HDa 20 mg every 24 hours
⬍60 kg: 30 mg twice daily 26 to 50 15 mg every 12 hours
10 to 25 15 mg every 24 hours
CAPD or HDa 15 mg every 24 hours
b
Tenofovir (Viread) 300 mg/d 30 to 49 300 mg every 48 hours
10 to 29 300 mg twice weekly
ESRD 300 mg weekly
CAPD or HDa 300 mg weekly
Zidovudine (Retrovir) 300 mg twice daily ⬍15 300 mg every 24 hours
CAPD or HDa 300 mg every 24 hours
Combination
medications
abacavir ⫹ 1 tablet daily Not recommended for
lamivudine CrCl ⬍50
(Epzicom)
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 233

Table 7. Continued
Antiretroviral Agent Daily Dosage CrCl (ml/min) Dosage
tenofovir ⫹ 1 tablet daily 30 to 49 1 tablet every 48 hours
emtricitabine
(Truvada)b
⬍30 Not recommended
efavirenz ⫹ 1 tablet daily Not recommended for
tenovovir ⫹ CrCl ⬍50
emtricidabine
(Atripla)b
zidovudine ⫹ 1 tablet twice daily Not recommended for
lamivudine CrCl ⬍50
(Combivir)
zidovudine ⫹ 1 tablet daily Not recommended for
lamivudine ⫹ CrCl ⬍50
abacavir (Trizivir)
Dosage adjustments for renal insufficiency are not necessary for nonnucleoside reverse transcriptase inhibitors (delavirdine, efavirenz, etravirine, and nevirapine),
protease inhibitors (atazanivir, darunavir, fosamprenavir, indinavir, lopinavir/ritonavir, nelfinavir, ritonavir, saqinavir, and tipranavir), CCR5 antagonists (maraviroc), fusion
inhibitors (enfuvirtide), or integrase inhibitors (raltegravir). CAPD, chronic ambulatory peritoneal dialysis; CrCl, creatinine clearance; eGFR, estimated GFR; HD,
hemodialysis.
a
Dose after HD.
b
Some experts recommend against prescribing for eGFR ⬍60 ml/min per 1.73 m2.
Reprinted from Advances in Chronic Kidney Disease, Vol 17, Kalayjian, RC, The treatment of HIV-associated nephropathy, pages 59 –71 (reference 17), Copyright 2010,
with permission from Elsevier.

19. Panel on Antiretroviral Guidelines for Adults and Adolescents: tors, and noradrenergic/serotonergic agonists, is pre-
Guidelines for the use of antiretroviral agents in HIV-1-infected
adults and adolescents, Washington, DC, Department of Health
sented in Table 9 (4). The SSRIs inhibit serotonin
and Human Services, Dcember 1, 2009. Available at: http://www.aidsinfo. reuptake, leading to increased serotonin levels in the
nih.gov/ContentFiles/AdultandAdolescentGL.pdf. Accessed March intersynaptic region. Of all antidepressants prescribed
1, 2010
to patients with CKD, these agents have the most
Psychiatric Medications clinical data available in the literature and with the
exception of paroxetine require no dosage adjustment
Antidepressant Drugs in renal impairment. They are generally well tolerated
Depressive illness is prevalent, affecting up to but can display common adverse effects, including
one third of patients with chronic kidney disease
sedation, headache, sexual dysfunction, insomnia,
(CKD) by some estimates (1,2). That few clinical data
nausea, and dry mouth. SSRIs display two unexpected
exist on antidepressant therapy in CKD underscores
adverse effects that frequently are managed by neph-
the observation that depression is underdiagnosed and
rologists: Hyponatremia as a result of the syndrome of
undertreated in this population (3). One might assume
that currently available antidepressant medications inappropriate antidiuretic hormone secretion (5) and
that are prescribed in the general population would be increased upper gastrointestinal (GI) tract bleeding in
efficacious in patients with CKD, yet such assump- patients who take concomitant nonsteroidal anti-in-
tions must be tempered by the realization that adverse flammatory drugs (NSAIDs) (6). The increased risk
drug effects are more frequent in this patient popula- for GI bleeding that is associated with SSRIs is
tion and efficacy less frequent as a result of altered thought to be related to intraplatelet serotonin defi-
pharmacokinetic and pharmacodynamic properties, re- ciency as a result of reduced reuptake, resulting in
spectively. inadequate serotonin release, which diminishes plate-
A limited review of so-called second-generation let aggregation (6). In the general population, the
antidepressants, including selective serotonin reuptake relative risk for GI tract bleeding associated with
inhibitors (SSRIs), serotonin norepinephrine reuptake concomitant SSRI/NSAID use (12.2 to 15.6) seems to
inhibitors, norepinephrine dopamine reuptake inhibi- be synergistic in that it exceeds the additive value of
234 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

Table 8. Major nephrotoxicities of medications used in HIV


Medication Reported Nephrotoxicity
NRTIs
zidovudine (AZT) None
lamivudine (3TC) Fanconi
emtricitabine (FTC) None
stavudine (d4T) None
abacavir (ABC) Fanconi, NDI
didanosine (ddI) Fanconi, NDI
tenofovir (TDF) ARF, Fanconi, NDI, hypophosphatemic osteomalacia
NNRTIs
nevirapine Acute kidney injury in the setting of DRESS
efavirenz Minimal change disease, urolithiasis, ARF
etravirine None
Protease inhibitors
nelfinavir Urolithiasis
amprenavir Urolithiasis
saquinavir Urolithiasis
lopinavir/ritonavir Urolithiasis
indinavir Urolithiasis, interstitial nephritis, ARF, papillary necrosis, NDI
atazanavir Urolithiasis, AIN
Entry inhibitors
enfuvirtide MPGN
maraviroc, vicriviroc None
integrase inhibitors
raltegravir None
elvitegravir None
AIN, acute interstitial nephritis; ARF, acute renal failure; DRESS, drug rash with eosinophilia and systemic symptoms; MPGN, membranoproliferative glomerulonephritis;
NDI, nephrogenic diabetes insipidus.
Reprinted from Advances in Chronic Kidney Disease, Vol 17, Jao J, Wyatt CM, Antiretroviral medications: adverse effects on the kidney, pages 72– 82 (reference 18,
Copyright 2010, with permission from Elsevier.

relative risk of each drug (2.6 to 3.6 for SSRIs, 3.7 to not discussed and should generally be avoided in
4.5 for NSAIDS) (6). Use of any antidepressant that patients with ESRD because of an increased risk for
increases serotonin levels requires close monitoring conduction abnormalities, arrhythmias, hyperthermia,
for signs/symptoms of serotonin syndrome, including and anticholinergic adverse effects. Anticholinergic
agitation, autonomic instability, confusion, diarrhea, adverse effects are important for nephrologists to be
dizziness, diaphoresis, hyperreflexia, hyperthermia, aware of, because urinary retention is one of the most
tremor, and unsteady gait, which may be seen after common and may lead to worsening kidney function
initiation of therapy or an increase in dosage (7). acutely.
Conversely, given the high rate of medication non- Antipsychotic medications with the exception of
compliance seen in the ESRD population, symptoms lithium are not presented in detail because such agents
such as nausea, insomnia, imbalance, hyperarousal, should be prescribed by psychiatrists and pharmacists
and malaise can be seen when serotonergic agents are who are working jointly as the primary treatment team
discontinued abruptly (“discontinuation syndrome”) and are beyond the scope of this issue of NephSAP;
(8). Last, nearly all antidepressants are metabolized by however, one life-threatening adverse effect associ-
the cytochrome P450 system, leading to numerous ated with antipsychotic, or “neuroleptic,” agents
drug– drug interactions (see references in Table 1). should be mentioned. Neuroleptic malignant syn-
Older agents for treatment of depression, such as drome is characterized by rigidity, bradyreflexia, hy-
tricyclic antidepressants (including amitriptyline, de- perthermia, autonomic instability, and mental status
sipramine, doxepin, imipramine, and nortriptyline) are changes and is seen with dopamine antagonism (9).
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 235

Table 9. Antidepressant agents (4)


Dosage Reduction for Ccr
Agent Adverse Eventsa <30 ml/min ⴙ ESRD
SSRI
citalopram Increased bleeding, SIADH None
escitalopram Increased bleeding, SIADH None
fluoxetine Increased bleeding, SIADH None
paroxetine Increased bleeding, SIADH Yes
sertraline Increased bleeding, SIADH None
Serotonin-norepinephrine reuptake inhibitor
duloxetine SIADH, hypertension Avoid
venlafaxine Hypertension (dosage related) Yes
Norepinephrine dopamine reuptake inhibitor
bupropion Seizures Yes
Other
mitazapine Edema Yes
SIADH, syndrome of inappropriate antidiuretic hormone secretion.
a
Urinary retention is a common adverse event with all agents.
Reprinted from Nephrology Nursing Journal, 2008, Volume 35, Number 3, pp 257–263 (reference 4). Reprinted with permission of the publisher, the American
Nephrology Nurses’ Association (ANNA), East Holly Avenue, Box 56, Pitman, NJ 08071-0056; 856-256-2320; FAX 856-589-7463; E-mail: nephrologynursing@ajj.com; Web
site: www.annanurse.org.

Lithium epithelial sodium channel from allowing lithium to


Used widely since the 1940s as an effective enter the principal cell and has demonstrated reversal
treatment for bipolar and major depressive disorders, of polyuria and isosthenuria (15). Whether long-term
lithium therapy may also be beneficial in delaying amiloride therapy, started early in a patient’s lithium
progression of amyotrophic lateral sclerosis and effec- regimen, can prevent the chronic tubulointerstitial in-
tive in Alzheimer’s disease, thus widening its thera- jury that is incurred by lithium exposure remains to be
peutic spectrum (10,11). A recent review of lithium seen. Nephrologists are often asked to see lithium-
nephrotoxicity by Grunfeld and Rossier (12) discussed treated patients in consultation for a variety of signs
the tubulointerstitial injury that is caused by lithium and symptoms that are associated with nephrogenic
and the consequent nephrogenic diabetes insipidus that diabetes insipidus, including polyuria and hypernatre-
results from downregulation of aquaporin 2 water mia; acute renal failure with concomitant angiotensin-
channel expression. After undergoing glomerular fil- converting enzyme inhibitor use (16) and azotemia as
tration and significant proximal tubular resorption (up a result of chronic impairment of renal function. Im-
to 80%), only a small amount of filtered lithium enters portant drug interactions that can lead to an abrupt rise
the distal nephron and undergoes further resorption via in serum lithium levels and acute lithium intoxication
the epithelial sodium channel on the apical membrane include concomitant use of diuretics (particularly thia-
of the collecting duct principal cell. Nonetheless, toxic zides) (17), NSAIDs (18), and angiotensin-converting
intracellular lithium levels may arise and are associ- enzyme inhibitors/angiotensin receptor blockers (19).
ated with inhibition of glycogen synthase kinase 3 In addition, management of lithium-induced renal tu-
␤-signaling pathways, which may be involved in the bular acidosis, hypercalcemia, hypothyroidism, and
maintenance of primary cilia (13). It is interesting that intoxication that requires acute dialysis therapy man-
the tubular microcystic transformation that is accom- dates consultation with a nephrologist. Signs and
panied by interstitial fibrosis and FSGS-type lesions in symptoms of lithium intoxication (both chronic and
lithium-treated patients may mirror the same micro- acute) as well as the indications for dialysis interven-
cystic changes that are seen in inherited cystic kidney tions can be found in dialysis and critical care hand-
diseases (e.g., autosomal dominant polycystic kidney books and are not discussed here. Numerous review
disease), in which primary cilia abnormalities have articles that discuss lithium and the kidney are avail-
recently been implicated (14). Amiloride blocks the able (20).
236 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

References 18. Reimann IW, Frolich JC: Effects of diclofenac on lithium kinetics.
1. Tossani E, Cassano P, Fava M: Depression and renal disease. Semin Clin Pharmacol Ther 30: 348 –352, 1981
Dial 18: 73– 81, 2005 19. Vipond AJ, Bakewell S, Telford R, Nicholls AJ: Lithium toxicity.
2. Hedayati SS, Minhajuddin AT, Toto RD, Morris DW, Rush AJ: Anesthesia 51: 1156 –1158, 1996
Prevalence of major depressive episode in CKD. Am J Kidney Dis 54: 20. Gitlin M: Lithium and the kidney: An updated review. Drug Saf 20:
424 – 432, 2009 231–243, 1999
3. Kimmel PL, Peterson RA: Depression in patients with end-stage
renal disease treated with dialysis: Has the time to treat arrived? Clin
J Am Soc Nephrol 1: 349 –352, 2006 Anticonvulsant Drugs
4. Raymond CB, Wazny LD, Honcharik PL: Pharmacotherapeutic op-
tions for the treatment of depression in patients with chronic kidney Older Anticonvulsant Drugs
disease. Nephrol Nurs J 35: 257–263, 2008
5. Jacob S, Spinler SA: Hyponatremia associated with selective seroto-
Despite their categorical name, anticonvulsant
nin-reuptake inhibitors in older adults. Ann Pharmacother 40: 1618 – drugs (ACDs) are used to treat an array of conditions
1622, 2006 other than seizure disorders, including pain associated
6. Mort JR, Aparasu RR, Baer RK: Interaction between selective sero-
tonin reuptake inhibitors and nonsteroidal antiinflammatory drugs:
with peripheral neuropathy (gabapentin and pregaba-
Review of the literature. Pharmacotherapy 26: 1307–1313, 2006 lin) and dementia-associated agitation (valproic acid).
7. Boyer EW, Shannon M: The serotonin syndrome. N Engl J Med 352: The use of such agents for patients with chronic
1112–1120, 2005
8. van Geffen EC, Hugtenburg JG, Heerdink ER, van Hulten RP, kidney disease (CKD) was reviewed by Israni et al.
Egberts AC: Discontinuation symptoms in users of selective seroto- (1), who broadly categorizes them into two groups:
nin reuptake inhibitors in clinical practice: Tapering versus abrupt Those that were available before the 1990s (“older”)
discontinuation. Eur J Clin Pharmacol 61: 303–307, 2005
9. Adnet P, Lestavel P, Krivosic-Horber R: Neuroleptic malignant and those that were developed more recently
syndrome. Br J Anaesth 85: 129 –135, 2000 (“newer”). Newer anticonvulsants were developed to
10. Fornai F, Longone P, Cafaro L, Kastsiuchenka O, Ferrucci M, Manca display more desirable pharmacokinetic properties
ML, Lazzeri G, Spalloni A, Bellio N, Lenzi P, Modugno N, Siciliano
G, Isidoro C, Murri L, Ruggieri S, Paparelli A: Lithium delays such as rapid and complete absorption from the gas-
progression of amyotrophic lateral sclerosis. Proc Natl Acad Sci trointestinal tract (100% bioavailability), linear kinet-
U S A 105: 2052–2057, 2008
ics (predictable drug concentrations at a given dos-
11. Martinez A, Perez DI: GSK-3 inhibitors: A ray of hope for the
treatment of Alzheimer’s disease? J Alzheimers Dis 15: 181–191, age), fewer drug– drug interactions, and less reliance
2008 on hepatic metabolism. As such, many of the newer
12. Grunfeld JP, Rossier BC: Lithium nephrotoxicity revisited. Nat Rev
Nephrol 5: 270 –276, 2009
compounds rely on renal excretion for elimination
13. Thoma CR, Frew IJ, Krek W: The VHL tumor suppressor: Riding from the body and thus warrant dosage reduction in
tandem with GSK3beta in primary cilium maintenance. Cell Cycle 6: the setting of renal impairment, as shown in Table 10.
1809 –1813, 2007
14. Gunay-Aygun M: Liver and kidney disease in ciliopathies. Am J Med
Of the older ACDs, phenytoin highlights the
Genet 151C: 296 –306, 2009 complex nature of altered pharmacologic properties
15. Batlle DC, von Riotte AB, Gaviria M, Grupp M: Amelioration of found in the setting of CKD. Phenytoin is highly
polyuria by amiloride in patients receiving long-term lithium therapy.
N Engl J Med 312: 408 – 414, 1985
protein bound (⬎90%), with only the unbound, or
16. Lehmann K, Ritz E: Angiotensin-converting enzyme inhibitors may “free,” fraction (⬍10%) demonstrating pharmacologic
cause renal dysfunction in patients on long-term lithium treatment. activity. In the uremic milieu of advanced kidney
Am J Kidney Dis 25: 82– 87, 1995
17. Jefferson JW, Kalin NH: Serum lithium levels and long-term diuretic disease, however, waste products displace phenytoin
use. JAMA 241: 1134 –1136, 1979 from its binding protein, resulting in a greater free

Table 10. Newer anticonvulsant drugs


Drug Dosage Reduction for Ccr <50 ml/min Supplement Dose after HD
Gabapentin Yes (CNS toxicity) Yes
Felbamate Yes Avoid
Lamotrigine No Unknown
Levetiracetam Yes Yes
Oxcarbazepine Avoid (hyponatremia) Avoid
Pregabalin Yes Yes
Topiramate Yes (renal calculi; RTA) Avoid (renal calculi)
Zonisamide Yes (renal calculi; RTA) Avoid (renal calculi)
CNS, central nervous system; HD, hemodialysis; RTA, renal tubular acidosis.
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 237

fraction. As such, one might be tempted to decrease tion, particularly those with severe impairment, unless
the dosage of phenytoin for patients with advanced dosage reduction occurs (8). Gabapentin toxicity is
CKD, but this would be incorrect. That more of the characterized by CNS symptoms including nystagmus,
drug is unbound means that more is available to ataxia, tremor, somnolence, and even coma in extreme
undergo hepatic metabolism, increasing clearance of cases; use of hemodialysis to remove excess drug can
the drug (2). As a result, the dosage of phenytoin is reverse these symptoms (9). Pregabalin is used for
generally not altered for dialysis patients, but it is seizure disorders, fibromyalgia, diabetic peripheral
imperative to monitor free phenytoin levels (therapeu- neuropathy, and postherpetic neuralgia. Like gabapen-
tic range 1 to 2 mg/L), not “total” phenytoin levels tin, pregabalin is minimally protein-bound and elimi-
(typically 10 to 20 mg/L), in this patient population. nated by the kidney with over 90% appearing un-
Because phenytoin is metabolized by hepatic P450 changed in the urine thus necessitating dose reduction
enzymes, concurrent use of medications that either in CKD (10). Hemodialysis significantly reduces pre-
induce or inhibit this enzyme system may lead to gabalin levels. Levetiracetam like gabapentin and pre-
subtherapeutic or toxic phenytoin levels, respectively, gabalin is not highly protein-bound and not hepatically
and should trigger increased drug monitoring of free metabolized. It undergoes glomerular filtration and
phenytoin levels. Fosphenytoin, a phenytoin prodrug thus its dose must be reduced in severe CKD patients
administered intravenously, can further complicate (11). It is also significantly removed by dialysis and
care because its metabolite can cross-react with phe- thus should be administered following dialytic therapy
nytoin assays, leading to falsely elevated levels (3). (11). Topiramate (12) and zonisamide (13) both inhibit
Hyponatremia is a frequent complication of car- carbonic anhydrase and thus are associated with renal
bamazepine therapy, although it is poorly understood tubular acidosis, metabolic acidosis, and renal stone
mechanistically (4). It may be caused by increased formation. Topiramate like other new ACDs is more
vasopressin release or a more robust vasopressin re- dependent on the kidney for elimination and dosage
ceptor effect at the level of the collecting duct or reduction is warranted when GFR is below 30 ml/min;
resetting of the hypothalamic osmoreceptors. Valproic it may also require supplemental dosing after dialysis
acid, like phenytoin and carbamazepine, is highly (12). Conversely, zonisamide demonstrates pharmaco-
protein bound and metabolized by the liver and under- logic parameters more in line with older ACDs in that
goes little renal elimination. All three agents have it is metabolized by the liver and moderately protein-
been associated with interstitial nephritis. Fanconi bound with only a third of a dose appearing in the
syndrome and hyponatremia have also been linked to urine unchanged and dosage reduction not generally
valproic acid use, and, rarely, hepatic failure has been indicated (13). Oxcarbazepine is structurally related to
reported (5). Phenobarbital has been linked to mega- carbamazepine and is associated with an even higher
loblastic anemia and osteomalacia, which may be incidence of hyponatremia (14). Unlike carbamaz-
difficult to interpret given the accompanying bone epine, the parent compound and active metabolite
disease and anemia seen with CKD. Some dosage undergo renal elimination mandating dose reduction in
reduction of phenobarbital may be in order with ad- the setting of renal impairment (15). Felbamate dis-
vanced CKD, because roughly 25% of a given dose plays a narrow therapeutic window with aplastic ane-
undergoes renal elimination (6). Ethosuximide is note- mia and hepatic failure complicating its use (16). Up
worthy because of its potential for causing drug- to 50% appears unchanged in the urine such that dose
induced systemic lupus erythematosus, including lu- reduction in advanced CKD is suggested (17). Both
pus nephritis (7). oxcarbazepine and felbamate affect the cytochrome
P450 enzyme system, however, thus complicating
Newer ACDs concomitant use with older ACDs and other hepati-
Gabapentin is used for seizure disorders, spastic- cally metabolized compounds. Lamotrigine like older
ity, and peripheral neuropathy pain. Like most of the ACDs is metabolized in the liver to an inactive me-
newer ACDs, it is not significantly protein-bound nor tabolite and not significantly removed by dialysis (18).
metabolized by the liver and is eliminated from the Nonetheless, therapeutic concentrations vary widely
body by glomerular filtration. As such, drug accumu- between patients, adverse effects do not appear to be
lation will occur in patients with reduced renal func- concentration-dependent and thus hard to predict, and
238 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

pharmacokinetic parameters may be altered with se- purposes and cooking, is increasingly popular in cap-
vere kidney impairment (18). Similar to older ACDs, sular form as a “natural” cholesterol-reducing agent.
interstitial nephritis has also been reported with lam- As with other “statins,” however, myopathy and rhab-
otrigine (Table 10). domyolysis have occurred in patients who take red
yeast rice, and a mycotoxin, citrinin, found in these
“Traditional” Medications, Food Contaminants, products may cause acute renal failure (22).
and Others A recent case report of lead-induced nephropathy
The term “traditional” medicine encompasses a as a result of ingestion of Ayurvedic herbal remedies
wide array of products that are alternatively referred to from India by a patient in Toronto, Canada, under-
as “herbal,” “natural,” “supplemental,” “nutritional,” scores the “global” aspect of nephrotoxic injury asso-
“organic,” “alternative,” “complementary,” and other ciated with unregulated products (23). Another exam-
labels. Use of such agents is growing worldwide ple of nephrotoxic injury in an era of globalization
(exceeding 80% in some populations) and as a result involves melamine, which first appeared in tainted
of nephrotoxic potential accounts for as much as one animal feed and then in contaminated milk-based in-
third of acute kidney injury where usage is common. fant formula, both manufactured in China but distrib-
Most of these compounds are produced outside con- uted worldwide. Melamine nephrotoxicity seems to
ventional regulatory pathways and as such are prone to stem from precipitation of melamine-cyanuric acid
contamination with additional toxins (e.g., L-trypto- crystals in the distal tubule and renal papillae with
phan contaminant resulting in eosinophilia-myalgia chronic inflammation, tubular necrosis, and stone for-
syndrome) (19). mation resulting, although much of what is know
Renal syndromes that result from exposure to comes from animal data (24). Melamine nephropathy
such agents reflect the area of the kidney damaged, can be added to the long list of crystal-induced/lithogenic
including glomerular injury (nephrotic syndrome renal diseases that are associated with both prescription
and thrombotic microangiopathy), tubular injury and nonprescription medications, including herbal/tradi-
(electrolyte wasting, renal tubular acidosis, concen- tional compounds, as listed in Table 11.
trating/diluting defects, and acute tubular necrosis),
hemodynamic disruption (acute renal failure), and Use of Extracorporeal Measures to Remove
parenchymal injury (crystal nephropathy, nephroli- Drugs and Chemicals
It is beyond the scope of this issue of NephSAP
thiasis, and obstruction). Nephrologists must be
to present an exhaustive discussion or comprehensive
aware of patient access to such products and include
list of all compounds that are removed by extracorpo-
exposure to these potential toxins in their differen-
real methods, but broad categories include pharmaceu-
tial diagnoses. Luyckx and Naicker (20) reviewed
the renal pathology of many traditional medicines
Table 11. Agents associated with crystal nephropathy
that are associated with kidney injury, and more
extensive lists can be found in standard references Melamine-cyanuric acid
(Table 1). A variety of nephrotoxic compounds are Silica-containing “supplements” (27)
Star fruit, Ma Huang, rhubarb, Djenkol beans, cranberry
discussed briefly.
juice
Aristolochic acid (AA) ingestion can lead to AA Pseudoephedrine (28)
nephropathy (previously termed Chinese herb nephrop- Vitamin C
athy or simply herbal nephropathy), which is character- Aspirin
ized by tubulointerstitial fibrosis, chronic renal failure Acycolovir
leading to ESRD, and urothelial cancer. The Aristolo- Methotrexate
chia species was mistakenly used in Chinese slimming Indinavir
agents instead of the less toxic Stephania species Foscarnet
Sulfonamide
because they share a common herbal name, Fang Ji
Triamterene
(20). Animal models suggested a potential role for Ciprofloxacin
hepatic P450 enzymes in the detoxification process of Ampicillin
AA (21). Cephalexin
Red yeast rice, long used for Chinese medicinal Adapted from references 26 –28.
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 239

Table 12. Properties of compounds that are removed by extracorporeal clearance methods
HD HP HF
Small molecule (⬍500 Da) Adsorbed by AC Small/midsize (up to 40,000 Da)
Small Vd (⬍1L/kg) Small Vd Small Vd
Minimally protein bound Minimally protein bound Higher % protein bound
Low endogenous clearance Low endogenous clearance Low endogenous clearance
Water soluble – –
AC, activated charcoal; HD, hemodialysis; HF, hemofiltration; HP, hemoperfusion.

tical agents as well as inorganic metals, solvents, 10. Bockbrader HN, Hunt T, Strand J, Posvar E, Sedman A: Pregabalin
pharmacokinetics and safety in healthy volunteers [Abstract]. Neu-
alcohols, herbicides, insecticides, and plant or animal rology 54: A421, 2000
venom. Extracorporeal methods to enhance elimina- 11. Patsalos PN: Clinical pharmacokinetics of levetiracetam. Clin Phar-
tion of substances from the body have included dial- macokinet 43: 707–724, 2004
12. Stowe CD, Bollinger T, James LP, Haley TM, Griebel ML, Farrar
ysis, hemoperfusion, and hemofiltration. It may be HC 3rd: Acute mental status changes and hyperchloremic metabolic
helpful to know basic principles that pertain to each of acidosis with long-term topiramate therapy. Pharmacotherapy 20:
these methods so that effective removal of toxic sub- 105–109, 2000
13. Faught E: Review of United States and European clinical trials of
stances can be achieved rapidly and cost-effectively. zonisamide in the treatment of refractory partial-onset seizures.
Table 12 outlines differences among these techniques. Seizure 135: S59 –S65, 2004
Hemoperfusion involves passage of blood through an 14. Sachdeo RC, Wasserstein A, Mesenbrink PJ, D’Souza J: Effects of
oxcarbazepine on sodium concentration and water handling. Ann
extracorporeal column that contains activated charcoal Neurol 51: 613– 620, 2002
to which certain compounds may adsorb. Previously 15. Rouan MC, Lecaillon JB, Godbillon J, Menard F, Darragon T, Meyer
used for theophylline, procainamide, or carbamaz- P, Kourilsky O, Hillion D, Aldigier JC, Jungers P: The effect of renal
impairment on the pharmacokinetics of oxcarbazepine and its metab-
epine overdose, hemoperfusion has largely been re- olites. Eur J Clin Pharmacol 47: 161–167, 1994
placed by high-flux/high-efficiency dialysis and/or he- 16. Johannessen SI, Battino D, Berry DJ, Bialer M, Krämer G, Tomson
T, Patsalos PN: Therapeutic drug monitoring of the newer antiepi-
mofiltration. Information regarding drug or toxin
leptic drugs. Ther Drug Monit 25: 347–363, 2003
removal can be readily obtained from poison control 17. Glue P, Sulowicz W, Colucci R, Banfield C, Pai S, Lin C, Affrime
centers, toxicology departments, and online resources MB: Single-dose pharmacokinetics of felbamate in patients with
renal dysfunction. Br J Clin Pharmacol 44: 91–93, 1997
as well as textbooks such as Medical Toxicology (25). 18. Fillastre JP, Taburet AM, Fialaire A, Etienne I, Bidault R, Singlas E:
Pharmacokinetics of lamotrigine in patients with renal impairment:
References Influence of haemodialysis. Drugs Exp Clin Res 19: 25–32, 1993
1. Israni RK, Kasbekar N, Haynes K, Berns JS: Use of antiepileptic 19. Slutsker L: Eosinophilia-myalgia syndrome associated with exposure
drugs in patients with kidney disease. Semin Dial 19: 408 – 416, 2006 to tryptophan from a single manufacturer. JAMA 264: 213–217, 1990
2. Liponi DF, Winter ME, Tozer TN: Renal function and therapeutic 20. Luyckx VA, Naicker S: Acute kidney injury associated with the use
concentrations of phenytoin. Neurology 34: 395–397, 1984 of traditional medicines. Nat Clin Pract Nephrol 4: 664 – 671, 2008
3. Annesley TM, Kurzyniec S, Nordblom GD, Buchanan N, Pool W, 21. Xiao Y, Ge M, Xue X, Wang C, Wang H, Wu X, Li L, Liu L, Qi X,
Reily M, Talaat R, Roberts WL: Glucuronidation of prodrug reactive Zhang Y, Li Y, Luo H, Xie T, Gu J, Ren J: Hepatic cytochrome
site: Isolation and characterization of oxymethylglucuronide metab- P450s metabolize aristolochic acid and reduce its kidney toxicity.
olite of fosphenytoin. Clin Chem 47: 910 –918, 2001 Kidney Int 73: 1231–1239, 2008
4. Dong X, Leppik IE, White J, Rarick J: Hyponatremia from carbam- 22. Prasad GV, Wong T, Meliton G, Bhaloo S: Rhabdomyolysis due to
azepine and oxcarbazepine. Neurology 65: 1976 –1978, 2005 red yeast rice (Monascus purpureus) in a renal transplant recipient.
5. Smith GC, Balfe JW, Kooh SW: Anticonvulsants as a cause of Transplantation 74: 1200 –1201, 2002
Fanconi syndrome. Nephrol Dial Transplant 10: 543–545, 1995 23. Prakash S, Hernandez GT, Dujaili I, Bhalla V: Lead poisoning from
6. Hoikka V, Savalainen K, Alhaua EM, Sivenius J, Karjalainen P, an Ayurvedic herbal medicine in a patient with chronic kidney
Parvianinen M: Anticonvulsant osteomalacia in epileptic outpatients. disease. Nat Rev Nephrol 5: 297–300, 2009
Ann Clin Res 14: 129 –132, 1982 24. Bhalla V, Grimm PC, Chertow GM, Pao AC: Melamine nephrotox-
7. Takeda S, Koizumi F, Takazakura E: Ethosuximide-induced lupus- icity: An emerging epidemic in an era of globalization. Kidney Int 75:
like syndrome with renal involvement. Intern Med 35: 587–591, 1996 774 –779, 2009
8. Blum RA, Comstock TJ, Sica DA, Schultz RW, Keller E, Reetze P, 25. Seifert SA: Elimination enhancement. In: Medical Toxicology, 3rd
Bockbrader H, Tuerck D, Busch JA, Reece PA, et al.: Pharmacoki- Ed., edited by Dart RC, Philadelphia, Lippincott Williams & Wilkins,
netics of gabapentin in subjects with various degrees of renal func- 2004, pp 269 –280
tion. Clin Pharmacol Ther 56: 154 –159, 1994 26. Perazella MA: Crystal-induced acute renal failure. Am J Med 106:
9. Wong MO, Eldon MA, Keane WF, Türck D, Bockbrader HN, 459 – 465, 1999
Underwood BA, Sedman AJ, Halstenson CE: Disposition of gabap- 27. Flythe JE, Rueda JF, Riscoe MK, Watnick S: Silicate nephrolithiasis
entin in anuric subjects on hemodialysis. J Clin Pharmacol 35: after ingestion of supplements containing silica dioxide. Am J Kidney
622– 626, 1995 Dis 54: 127–130, 2009
240 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

28. Smith CL, Gemar SK, Lewis MJ: Pseudoephedrine urolithiasis asso- Basiliximab and Daclizumab
ciated with acute renal failure. Neprhol Dial Transplant 19: 263–264,
2004
In the past two decades, a greater understanding
of the immunobiological processes that are involved in
allograft rejection have led to significant discoveries in
Immunosuppressive Therapies the field of clinical transplantation and production of
humanized and chimeric antibodies. Compared with
Induction Therapy traditionally engineered mAbs or polyclonal antibod-
Polyclonal antibodies were the first class of bi- ies, these newer biological agents have not been asso-
ological agents used for the induction or treatment of ciated with cytokine release reactions and have been
acute cellular rejection. The discovery of hybridoma in associated with a very limited increased risk for op-
1975 provided a new direction to manufacture several portunistic infections (8,10).
different mAbs for diagnostic and therapeutic pur- During acute cellular rejection, T cells are stim-
poses (1). OKT-3 was the first monoclonal biological ulated. Activation of T cells and other cytokines re-
agent for the treatment of acute rejection. OKT-3 sults in activation of IL-2 receptor (IL-2R) ␣ chain
targets the CD3 complex expressed on mature T cell (CD25), which is located on the surface of antigen-
receptors (1,2). Biological agents that were found to be active T cells. IL-2 is an important cytokine that
effective in the treatment of acute rejection were then activates cytotoxic T cells and increases helper T cell
used for induction therapy in the early posttransplan- proliferation, magnifying the allograft rejection cas-
tation period (3,4). These agents provide a high and cade. In addition, IL-2 may affect the proliferation of
rapid level of immunosuppression in the vital early natural killer cells, macrophages and monocytes, B
posttransplantation period, when the risk for acute cells, epidermal dendritic cells, and lymphocyte-acti-
rejection is highest. Although induction therapy is not vated killer cells. Inhibition of IL-2 may ultimately
indicated for most patients, it is proved to decrease decrease inflammatory responses and allograft rejec-
acute rejection early after transplantation and improve tion (11,12).
long-term allograft survival rate in immunologically Basiliximab and daclizumab are engineered
high-risk kidney transplant recipients. Patients who IgG1 mAbs that bind to the ␣ chain of the IL-2R
(CD25), which is expressed on the surface of activated
benefit most from induction therapy include those with
lymphocytes (6 – 8). The IL-2R is made of three high-
preformed antibodies (panel-reactive antibodies ⬎20%),
affinity subunits: The ␣ subunit (TAC antigen or
previous organ transplants, delayed graft function,
CD25), the ␤ subunit (CD122), and the ␥ subunit
HLA mismatches, multiorgan transplantation, and pro-
(CD132). For transduction of an inflammatory signal
longed cold ischemic time and patients with expanded-
or activate T cells, ␣ subunit and ␤ subunit binding is
criteria donors (5). More important, the use of biolog-
necessary. Daclizumab and basiliximab exert their ef-
ical agents early after transplantation allow for a delay
fects by binding to the ␣ subunit of the IL-2R, thereby
in the use of calcineurin inhibitors (CNIs) to avoid acting as a competitive inhibitor of IL-2 (13–15).
associated nephrotoxicity (6). Induction therapy with Daclizumab is a humanized antibody in which
biological agents with T cell– depleting properties (an- portions of the variable region that are not associated
tilymphocyte antibodies, OKT-3, the antithymocyte with the binding site are replaced by human amino
antibodies, and alemtuzumab) may also treat early acid sequences to decrease immunogenicity; therefore,
subclinical acute rejection or clinical acute rejection ⬎90% of the antibody is human IgG, and only hyper-
during delayed graft function, a time when renal func- variable regions are mouse antibody sequences. In
tion cannot be assessed correctly via computation of contrast, basiliximab is a chimeric antibody in which
urine output and elevation of serum creatinine or other the murine constant region of the Ig is replaced by
urinary biomarkers (7–9). Cytokine release syndrome human amino acid sequences. Basiliximab is con-
is a common complication of T cell– depleting agents structed by combining mouse variable regions with the
after the first two doses and requires high-dosage constant region of human IgG1 (13–15).
methylprednisolone (1 mg/kg), diphenhydramine, and The efficacy and safety of both daclizumab and
acetaminophen 30 to 60 minutes before infusion and basiliximab have been proved in a number of clinical
every 4 to 6 hours as needed (2,3). studies. Compared with placebo, at 12 months, a
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 241

significant reduction in acute rejection was reported in cyte globulin is the preferential agent over equine
the daclizumab-treated group (43 versus 28%; P ⬍ antithymocyte globulin (19).
0.001); however, there was no statistical difference in After T cell lysis, there is a significant release of
patient and allograft survival at 3 years (16,17). cytokines. Release of cytokines after the first two
Basiliximab and daclizumab are well tolerated. doses is attributed to infusion-related reactions such as
In addition, administration of these agents has not fever and chills (70%); headache (40%); serum sick-
been associated with significant clinical toxicity or ness and back pain (40%); nausea, diarrhea, and diz-
cytokine release syndrome (fever, chills, headache, ziness (30%); and myelosuppression (leukopenia and
and pulmonary edema), which is commonly observed thrombocytopenia; 45%) (20). The common dosing
with T cell– depleting muromonab-CD3 therapy. Ad- strategy of thymoglobulin for the treatment of acute
verse effects that are observed among basiliximab- and rejection is 1 to 1.5 mg/kg per d for 7 to 10 days with
daclizumab-treated patients are similar to those of T cell monitoring. When used for induction therapy,
placebo-treated patients. No drug interactions have the dosage is 1.5 mg/kg for two to four doses. Equine
been reported with the use of basiliximab and dacli- antithymocyte globulin is administered at 10 to 15
zumab. The dosage of basiliximab is 20 mg intrave- mg/kg per d intravenously for 10 to 14 days for
nously and should be given before the transplant treatment of acute rejection and up to 7 days for
followed by a second 20-mg dose on day 4 after induction therapy. The first dose usually begins during
transplantation. the preoperative period or shortly after transplantation.
The Food and Drug Administration–approved For both equine antithymocyte globulin and rabbit
dosages of daclizumab are 1 mg/kg within 24 hours of antithymocyte globulin, the first two doses should be
transplant surgery and then 1 mg/kg administered given over 6 hours after administration of methylpred-
every 2 weeks after surgery for a total of five doses; nisolone intravenously, and acetaminophen and di-
however, most transplant protocols dictate a two doses phenhydramine should be given 30 to 60 minutes
with a preoperative dose and another dose at the time before the dose and as needed every 6 hours for fever,
of discharge (18). No dosage adjustment is necessary chills, and other infusion-related reactions. Compared
in renal impairment, but no data are available for with Atgam, which must be administered through a
dosage adjustments in hepatic dysfunction. Safety is central line, rabbit antithymocyte globulin can be
one of the most evident benefits of induction therapy given through a peripheral line with addition of hep-
with IL-2R antibodies compared with other induction arin and hydrocortisone to 500 ml of normal saline
agents. In 2010, Roche Pharmaceuticals and Diagnos- solution (21).
tics will discontinue commercialized daclizumab for Muronomab-CD3 (OKT-3)
organ transplant patients. Since introduction of rabbit antithymocyte glob-
Antithymocyte Globulin Equine/Rabbit ulin, the use of OKT-3 has been limited only to
Polyclonal antibodies such as equine and rabbit patients with acute cellular rejection that is resistant to
antithymocyte globulin react with a number of T cell other agents. CD3 is expressed on activated T cells,
surface receptors, including CD2, CD3, CD4, CD8, and OKT-3 is a murine mAb that targets the CD3
CD11a, CD25, CD40, and CD54. Polyclonal antibod- receptor. Webster et al. (22) reviewed 21 studies (49
ies exert their pharmacodynamic properties in three reports, 1387 patients) of the use of T cell– depleting
different ways. Initially, by binding to cell surface agents for the treatment of steroid-resistant rejection.
markers, antithymocyte globulin may promote cell There was no benefit of muromonab-CD3 over either
lysis through complement-mediated process. Second, equine or rabbit antithymocyte globulin with regard to
antithymocyte globulin promotes T cell depletion treatment of rejection, preventing subsequent rejec-
through opsonization by the reticuloendothelial sys- tion, or preventing graft loss or death. OKT-3 induces
tem. Finally, antithymocyte globulin provides immu- T cell depletion through antibody-dependent cell cy-
nosuppression through vital T cell surface molecule totoxicity (ADCC), cell coating, and antigenic modu-
(CD2, CD3) inversion and modulation. Although lation. Most likely, the ADCC and cell coating are
modulated cells are circulating, in general, these cells more important initially, whereas modulation is more
are inactive and dysfunctional. Because of greater important later. OKT-3 binds to the CD3 receptor
potency and ease of administration, rabbit antithymo- epitope of the T cell receptor portion of the T lym-
242 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

phocyte cell membrane. Coating this target molecule plications after alemtuzumab induction have varied with
prevents activation of T lymphocytes and facilitates the dosage that was used. Anemia, neutropenia, throm-
lymphocyte removal from circulation via opsonization bocytopenia, headache, infusion-related reactions, and
through ADCC (23,24). infections are common after administration of alemtu-
This agent is dosed at 2 to 5 mg/d for the zumab. Premedication with acetaminophen and oral an-
treatment of acute rejection or induction and usually is tihistamines is advisable to reduce the incidence of infu-
given for a total of 10 to 14 days. When used alone, sion-related reactions (26 –28).
OKT-3 may induce a rapid and strong human anti-
mouse antibody, which limits its use as an immuno- Maintenance Therapy
suppressive agent. The objectives of maintenance immunosuppres-
The major adverse effects of OKT-3 are flu-like sion are to prevent acute and chronic allograft rejec-
symptoms; fever, chills, nausea, vomiting, diarrhea, tion. Maintenance immunosuppression should mini-
and headache are most often observed after the first mize the risk for rejection while improving the quality
two doses. These reactions are related to release of of life and reduce the rate of allograft loss or death. To
cytokines from activated T lymphocytes and mono- balance the risk for rejection and infection/malig-
cytes. Fatal pulmonary edema has been observed nancy, drugs must be selected with vigilance and
among patients with fluid overload or in patients with dosage should be titrated according to plasma drug
uncompensated heart failure. Capillary leak syndrome concentration or the risk for adverse drug reactions. In
results from cytokine release and is thought to be a addition, optimized patient and graft survival can be
contributory factor to the development of pulmonary acquired through avoidance or discontinuation of
edema. Before use of OKT-3, the weight of the patient drugs with the highest risk for toxicities; however,
should be within ⱕ3% of minimum dry weight, and a sudden discontinuation of immunosuppression may
chest x-ray should be obtained within 24 hours to rule increase the risk for late rejection, which is a proven
out any evidence of pulmonary edema. OKT-3 has poor prognosis and increases the risk for chronic
also been associated with cytokine-induced nephrop- allograft nephropathy.
athy. Because of these adverse drug reactions and The most commonly used maintenance immuno-
safer T cell– depleting agents, the role of OKT-3 is suppressants include a double or triple protocol that
limited to the treatment of severe acute rejection consists of the following classes of drugs: CNIs (cy-
(23,24). closporine [CsA] and tacrolimus), antiproliferatives
Alemtuzumab (azathioprine and the mycophenolic acid [MPA] de-
Preliminary results from a number of single- rivatives), target of rapamycin (TOR) inhibitors
center studies suggested that alemtuzumab could be an (sirolimus and everolimus), and corticosteroids (meth-
important agent for prevention of allograft rejection. ylprednisolone and prednisone derivatives). Comorbid
Alemtuzumab is a recombinant DNA-derived mAb medical conditions as well as potential drug interac-
that binds to CD52 and causes profound lymphopenia. tions that may affect long-term graft survival or in-
Alemtuzumab is not approved in transplantation and is crease the risk for adverse drug reactions should be
used primarily for the treatment of B cell lymphocytic considered. For example, although it is not contrain-
leukemia. CD52 is located on the cell surface of B and dicated, for patients who have a history of gout and are
T lymphocytes, many macrophages, natural killer taking allopurinol, the use of azathioprine should be
cells, and a subpopulation of granulocytes. Alemtu- avoided or the dosage adjusted to 0.5 mg/kg per d.
zumab affects B and T cells through complement- Calcineurin Inhibitors
mediated or antibody-dependent cell lysis. When used Both CsA and tacrolimus belong to a class of
for induction therapy, after a single dose of 30 mg, immunosuppressants called CNIs (29). CsA and ta-
alemtuzumab produces extensive and prolonged im- crolimus bind to their specific immunophilins and
munosuppression (25). A study of the use of a single inhibit dephosphorylation of N-FAT molecules. The
30-mg dose preoperatively showed a low acute rejec- introduction of CNIs was associated with a significant
tion rate with a lower risk for infections in high-risk improvement in both patient and allograft survival at 1
recipients (21). year. CsA is a cyclic undecapeptide, which was dis-
The reported adverse reaction and long-term com- covered from fungus isolated from Norwegian soil
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 243

(Tolypocladium inflatum) (29). In 1972, CsA was patients. Other functional manifestations of CsA neph-
initially used in preclinical and clinical transplantation rotoxicity are hypomagnesemia, hyperuricemia, and
(30). CsA effects the activation and proliferation of T hypocalcemia, which are independent of changes in
lymphocytes through inhibition of IL-2 transcription. serum creatinine. Acute dosage-dependent nephrotox-
The oral absorption of CsA is slow, erratic, and in- icity associated with CsA is usually reversible within
complete, ranging from 10 to 70% with a mean bio- 1 week with appropriate dosage reduction (31,39).
availability of approximately 30%. Peak blood con- The most serious dosage-independent and irrevers-
centrations are usually achieved 2 to 6 hours after ible toxicity of CsA is a progressive chronic nephropa-
administration of the standard formulation. The plasma thy. Therapeutic drug monitoring has a very limited
concentration at 2 hours has much better correlation to correlation with chronic histopathologic changes (31,38).
the area under the curve (AUC) and clinical benefit of Some patients may develop ESRD and require renal
CsA when compared with 12-hour trough levels (31). replacement therapy. Kidney biopsy in patients with CsA
Most centers, however, still use 12-hour trough levels nephrotoxicity suggests afferent arteriolopathy and a
for CsA monitoring. Even though 2-hour levels corre- striped pattern of tubulointerstitial atrophy and fibro-
late with AUC much better than trough levels, 2-hour sis. Finally, hemolytic uremic syndrome and acute
plasma concentration monitoring is not practical for thrombotic thrombocytopenic purpura are significant
centers with many transplant patients (32). The ab- but rare adverse effects of CsA therapy.
sorption of CsA is subject to significant inter- and Although the exact mechanism of CsA-in-
intrapatient variability and can be affected by the duced hypertension has not been elucidated, hyper-
presence of food, fat content in the food, gastrointes- tension is a common problem after transplantation,
tinal motility, and diarrhea (33). For improvement of
even in patients with stable renal function. Although
the pharmacokinetic properties of CsA and decrease
hypertension is dosage and concentration depen-
inter- and intrapatient variability, a microemulsion
dent, it can be observed in patients with therapeutic
formulation of CsA (Neoral) was developed in the
or subtherapeutic plasma concentrations. Plasma
early 1990s. Despite better pharmacokinetic proper-
renin activity is usually normal or only mildly
ties, microemulsion CsA has not resulted in better
elevated in CsA-treated patients with hypertension,
patient or graft survival. The pharmacokinetics of CsA
suggesting that hypertension in these patients is
follows a zero-order kinetic model and is subject to a
mostly volume dependent (2,40).
significant enterohepatic redistribution (recycling)
(34). CsA is widely distributed throughout the body Other major adverse effects of CsA are listed in
and is highly tissue bound. The volume of distribution Table 13. Most of these adverse reactions can largely
ranges from an average of 3.5 to 13.0 L/kg. CsA is be managed by careful therapeutic drug monitoring.
extensively metabolized through the cytochrome P450 The most common oral adult dosage of CsA
IIIA4 isoenzyme system (35). The clearance of CsA is ranges from 3 to 5 mg/kg per d in two divided doses
higher in children compared with adults, and only 1% in transplant patients, whereas 1 to 3 mg/kg per d is
of the drug is excreted unchanged through the urine. common for most autoimmune diseases. When con-
Dialysis or renal dysfunction does not affect the phar- verting a patient from oral to intravenous administra-
macokinetics of CsA; therefore, CsA can be given tion, the dosage should be reduced to approximately
before or after any type of renal replacement therapy. one third of the oral dose and not exceed more than 2
Nephrotoxicity and hypertension are commonly re- mg/kg per d. A number of CsA assays (HPLC, thera-
ported with CsA (36,37). These adverse reactions are peutic drug monitoring, polyclonal, and monoclonal)
concentration dependent. CsA nephrotoxicity has been with different specificity for parent compounds or
observed in 20 to 30% of kidney, heart, and liver inactive metabolites exist, creating an associated range
transplant recipients (36,37). This nephrotoxicity is of therapeutic whole-blood concentrations. The type
particularly important when CsA is used in patients of assay, renal function, comorbid conditions, and
with marginal kidney function or in patients with time since transplantation all may affect desirable
delayed graft function (38). Hypertension, fluid reten- plasma concentrations after transplantation. The ther-
tion, hyperkalemia, hyperchloremia, and metabolic ac- apeutic range for more specific assays of CsA is
idosis are also frequently observed in CsA-treated between 150 and 250 ng/dl (33,41).
244 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

Table 13. CsA and tacrolimus drug– drug interactions


Drug Mechanism Effects Severity Comments
ACEIs Renal dysfunction in RAS 1 serum creatinine 3 Physiologic elevation of
creatinine; anemia;
class effects
Acyclovir Inter-renal crystallization Stone formation 4 Avoid dehydration;
infuse over 1 hour
Amiodarone 2 clearance Nephrotoxicity 3 Very slow onset and
offset
Amlodipine 2 clearance 1 CNI level 4 Nifedipine is preferred
dihydropiradine
Amphotericin B Synergistic nephrotoxicity Nephrotoxicity 3 Require hydration and
electrolyte monitoring
Carbamazepine 1 clearance 2 CNI level 3 Slow onset (up to 7
days)
Chloroquine 2 clearance 1 CNI level 3
Cholestyramine 1 clearance 2 CNI level 4 Separate doses by 3
hours
Ciprofloxacin Possible 2 CSA effects Pharmacodynamic 4 May increase risk for
on IL-2 antagonism rejection
Clarithromycin 2 clearance 1 CNI level 2 Azithromycin is the
preferred agent
Clotrimazole troches 2 clearance of TAC Increase TAC level 3 Monitor TAC level
closely; no effect on
CsA
Colchicine Synergistic toxicity Increase neurotoxicity 3 Gastrointestinal
dysfunction and
neuromyopathy
Co-trimoxazole Inhibit creatinine Increase serum creatinine 4 Preferred agent for
secretion pneumocystis
pneumonia
Digoxin CSA may 2 clearance of Increase digoxin level 3 Monitor digoxin level
digoxin closely
Diltiazem 2 clearance 1 CNI level 3 Can be used to raise
CNI level when
needed
Erythromycin 2 clearance 1 CNI level 2 Azithromycin is the
preferred agent
Fluconazole 2 clearance 1 CNI level 3 Increase liver function
tests also
Fluvoxamine 2 clearance 1 CNI level 2
Fosphenytoin 1 clearance 2 CNI level 3 Monitor levels carefully
HMG-CoA reductase 2 clearance of statins Myopathy; 3 Less effect with TAC;
inhibitors (statins) (greater effect with rhabdomyolysis pravastatin is
CSA) preferred because it
has the least
interaction
Itraconazole 2 clearance 1 CNI level 3 Monitor levels
carefully; decrease
dosage 50 to 85%
Ketoconazole 2 clearance 1 CNI level 3 Monitor levels
carefully; decrease
dosage 25 to 75%
Methylprednisolone 2 clearance 1 CNI level 3 Only at high dosages
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 245

Table 13. CsA and tacrolimus drug– drug interactions


Drug Mechanism Effects Severity Comments
Metoclopramide 2 gastric emptying time 1 CNI level 3 Increased peak and
AUC by 25 to 50%
NSAIDs/COX-2 Synergistic nephrotoxicity Nephrotoxicity 3 CNI induced
inhibitors vasoconstriction is
exacerbated by
prostaglandin
inhibition
Phenobarbital 1 CNI clearance 2 CNI level 3 Slow onset, slow offset
Phenytoin 1 CNI clearance 2 CNI level 3
Rifabutin 1 CNI clearance 2 CNI level 3 Rifabutin is a less
potent hepatic enzyme
inducer than rifampin
Rifampin 1 CNI clearance 2 CNI level 2
Verapamil 2 CNI clearance 1 CNI level 3 Anticipate decrease in
CNI dosage
1, Avoid combination; 2, usually avoid (use only no other alternative agents available); 3, monitor closely; 4, no action needed (the risk for adverse drug reaction is
small). ACEI, angiotensin-converting enzyme inhibitor; NSAID, nonsteroidal anti-inflammatory drug; RAS, renin-angiotensin system; TAC, tacrolimus.

Tacrolimus (FK-506) mus binds to a high-affinity intracellular FK-binding


Tacrolimus (Prograf, FK-506) is another CNI protein (FK-BP-12). The complex of FK-BP-12 and
produced from Streptomyces tsukubaensis, a fungal tacrolimus inhibits calcineurin, which interacts with a
organism found in Japanese soil (42,43). Tacrolimus calcium/calmodulin-dependent serine/threonine phos-
can be administered orally or by continuous intrave- phatase. Dephosphorylation is essential for activation
nous infusion and is approximately 20 to 25% bio- of N-FAT and activation of IL-2 transcriptions. Thus,
available when given orally, although this ranges from tacrolimus exerts its immunosuppressive properties
5 to 30%. The usual initial oral dosage for tacrolimus through inhibition of transcription of IL-2 genes (45).
ranges from 0.1 to 0.2 mg/kg per d in two divided Nephrotoxicity, hypertension, new-onset post-
doses. When converting a patient from oral to intra- transplantation diabetes, and neurotoxicity are the
venous administration, the dosage should be reduced most common adverse effects of tacrolimus. These
to approximately 15 to 20% of the total daily oral adverse reactions have been primarily observed at
dosage. Tacrolimus is highly bound to albumin, ␣-1 plasma concentrations of ⬎15 ng/dl but can occur at
acid glycoprotein, and erythrocytes. Changes in any plasma concentration. Diabetes usually occurs at
plasma protein binding can alter the pharmacokinetic plasma concentrations of ⬎10 ng/dl and with concom-
properties of tacrolimus. The usual therapeutic plasma itant use of prednisone. Nephrotoxicity from tacroli-
concentration of tacrolimus is 5 to 15 ng/ml (44). The mus is much less predictable than with CsA and can
concentration of tacrolimus is significantly higher in occur over a wide range of plasma concentrations but
whole blood compared with plasma concentration. generates a pattern of nephrotoxicity similar to CsA.
Thus, whole blood should be used for therapeutic drug Although less nephrotoxicity and fewer renal hemo-
monitoring of tacrolimus. Like CsA, tacrolimus is dynamic changes were initially reported with tacroli-
primarily metabolized in the liver via the cytochrome mus, more recent reports have documented a very
P450-IIIA4 system; however, tacrolimus is more sub- similar incidence of nephrotoxicity compared with
ject to important drug interactions compared with similarly therapeutic doses of CsA, with an overall inci-
CsA. dence of nephrotoxicity that approaches 35 to 40%. Like
Like CsA, tacrolimus is a prodrug and shares CsA, long-term administration of tacrolimus may cause
similar molecular targets even though tacrolimus is striped interstitial fibrosis and arteriolar hyalinosis. A
structurally dissimilar. Similar to CsA, the exact se- number of metabolic adverse reactions, such hyperkale-
quence of events that lead to the immunosuppressive mia, hyperuricemia, and hypomagnesemia, have also
action of tacrolimus has not been elucidated. Tacroli- been observed with the use of tacrolimus. Generally,
246 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

patients who receive tacrolimus require fewer antihy- kinetic behavior has been proved (rate, extent of ab-
pertensive and hyperlipidemic agents compared with sorption, AUC, and time to maximal concentration).
CsA-treated patients, but the overall incidence of new- Regulatory guidelines state that drug products are AB
onset diabetes is much higher with tacrolimus than rated when the 90% confidence interval for the ratio of
CsA. Alteration in peripheral insulin resistance and a the mean response is within 80 and 125%. This is
decrease in insulin release contribute to hyperglycemia different from variation of 80 to 125% of plasma
in these patients. Hirsutism and gingival hyperplasia, concentration that has been claimed by a number of
which frequently are observed in CsA-treated patients, pharmaceutical companies. The use of powerful sta-
are very rare and are not considered a true adverse tistical criteria and two one-sided tests does not allow
effect of tacrolimus (46 – 48). any drug product with a mean plasma difference and
Generic Formulations of CNIs AUC ⬎10% to be approved as an AB-rated agent. In
As the cost of health care continue to soar, it is a study of 2070 clinical bioequivalence studies of
vital to provide a cost-effective immunosuppressive generic drugs in the United States, the average differ-
regimen to provide adequate immunosuppression and ence in value of Cmax and AUC between generic and
avoid acute rejection while decreasing toxicity and innovator products was 4.3 and 3.5%, respectively
overall cost related to immunosuppressive agents. (55). Differences between branded drugs and generic
Since approval of the Hatch-Waxman Act in 1983, drugs can include variations only in shape, color, size,
generic drugs have provided four major benefits to the expiration date, and inactive ingredients; however,
US health care system: (1) Stimulate competition, (2) none of these factors should have any effect on drug
expand dollar expenditure for other health care ser- pharmacokinetics or pharmacodynamics of a specific
vices, (3) promote creation of smaller pharmaceutical agent.
companies, and (4) reduce prices (49 –53). Today,
although generic drugs supply 75% of the US market, Azathioprine
they account for only 10% of total drug expenditure Azathioprine is an imidazole analogue of 6-mer-
because generic drugs are typically priced at 10 to captopurine (6-MP) that inhibits both de novo and
50% of innovator products. Although there are some salvage pathways for T and B lymphocyte prolifera-
unproven concerns about the use of generic drugs, tion. Since introduction of mycophenolate mofetil
most data support the use of generic immunosuppres- (MMF) to the market, use of azathioprine has declined
sive agents for transplant patients. It is important for markedly, but it is still an excellent choice for patients
nephrologists to have an understanding of generic drug who are intolerant to MMF. After oral administration,
development, yet in a study of 3639 physicians nation- the bioavailability of azathioprine ranges from 50 to
wide, only 17% were able to recognize the Food and 60% with peak plasma concentration at 2 hours. Aza-
Drug Administration standards for bioequivalency for thioprine is distributed widely to all body compart-
generic drugs (54). ments with a volume of distribution that seems to
According to the Food and Drug Administration, correlate with total body weight. Liver failure may
drug products are considered AB rated when test alter the clearance of azathioprine, and dosage adjust-
results provide clinical evidence that generic drugs and ment is needed to avoid toxicity. Therapeutic drug
innovator agents are bioequivalent. AB-rated products monitoring for azathioprine has no clinical value as a
ensure safety and efficacy similar to innovator prod- result of the intracellular pharmacodynamic effects of
ucts and are virtually the same as the brand name azathioprine on thiopurine nucleotides (56,57).
product but with a lower price. For drug products to be Adverse reactions that are associated with aza-
regarded as bioequivalent, pharmaceutical equivalence thioprine therapy are relatively uncommon and are
needs to be demonstrated. Drugs are considered phar- rare with dosages of ⬍2 mg/kg per d. The most
maceutical equivalents when products have the same commonly encountered adverse event is myelosup-
active ingredient(s), dosage form, route of administra- pression. Dosage adjustment is critical for patients
tion, and strength and provide the same concentration with abnormal blood counts (white and red blood cells
at the receptor side. Drug products are considered and platelets) and in patients who take allopurinol,
bioequivalent when pharmaceutical equivalency has because allopurinol and other xanthine oxidase inhib-
been met and clinical evidence of similar pharmaco- itors may predispose patients to excessive bone mar-
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 247

row suppression. Azathioprine has also been associ- reversible when the drug is discontinued or dosage is
ated with veno-occlusive disease and pancreatitis. The decreased. Although the mechanism of this complica-
experience with azathioprine during pregnancy in kid- tion is unclear, it may be related to local or systemic
ney transplantation is encouraging, and compared with effects. Unfortunately, reduction in MPA dosage or
MMF, there is very limited risk for congenital anom- discontinuation of MPA for gastrointestinal toxicity is
alies with azathioprine (58); prednisone, CsA, tacroli- associated with a significant risk for graft failure
mus, and azathioprine have the lowest risk for con- (hazard ratio 2.36) (59).
genital malformations compared with other commonly After oral administration, MMF is rapidly con-
used immunosuppressive agents (Table 14). verted to MPA in the acidic environment in the stom-
MPA Derivatives ach. Unlike MMF, mycophenolate sodium is absorbed
MPA is a purine antagonist that was approved in in the lower gastrointestinal tract. The slow rate of
1995 for the prevention of acute rejection in renal absorption also results in a delayed peak plasma con-
transplant recipients. It is a noncompetitive inhibitor centration for mycophenolate sodium; the Cmax for
of inosine monophosphate dehydrogenase, an enzyme MMF occurs at 0.5 to 1.0 hours after oral administra-
that plays a vital role in the de novo pathway of purine tion, whereas for mycophenolate sodium, the serum
biosynthesis. Both T and B cell proliferation require peak concentration occurs at approximately 1.5 to 3.0
inosine monophosphate dehydrogenase for activation hours (60).
and maturation; therefore, mycophenolate inhibits the Because mycophenolate has shown a great phar-
rate-limiting enzymes in T and B cell biosynthesis. macokinetic variability in various patient populations,
MPA-based regimens have been effective at improv- routine mycophenolate therapeutic monitoring has
ing long-term allograft survival and reducing acute been suggested; however, data about the utility of
rejection of kidney transplant patients. Although it has therapeutic drug monitoring are inconclusive. In two
become a standard of practice to prescribe MPA-based separate randomized clinical trials of kidney transplant
therapy at the time of discharge, the result of clinical recipients, mycophenolate therapeutic drug monitor-
studies and safety of MPA-based therapy must be ing versus fixed dosage of mycophenolate did not
interpreted in the context of study design and dosage show any differences in biopsy-confirmed acute rejec-
of comparative agents. Different salt forms of myco- tion, mean serum creatinine concentrations, incidence
phenolate are on the US market: MMF (CellCept [F. of cytomegalovirus or polyomavirus infection, or al-
Hoffman La-Roche] and generic formulations) and lograft loss (60). A number of factors are known to
mycophenolate sodium (myFortic; Novartis). alter significantly the pharmacokinetic behavior of
The most common adverse effect associated with mycophenolate in kidney transplant recipients. Renal
mycophenolate is gastrointestinal toxicity, which is failure, hepatic dysfunction, choice of CNI, dosage of

Table 14. Azathioprine and mycophenolate drug– drug interactions


Drug Mechanism Effects Severity Comments
ACEIs Synergistic Anemia, neutropenia 3 1 bone marrow toxicity
myelosuppression
Acyclovir 1 AUC of MMF Not significant 4
Allopurinol Inhibits xanthene oxidase, Severe neutropenia 1 2 AZA dosage by 75%
1 AUC of AZA
Antacids 2 absorption of MMF 2 efficacy 3
Cholestyramine 2 absorption of MMF 3 1 bone marrow toxicity
Co-trimoxazole Synergistic Anemia, neutropenia 3
myelosuppression
Ganciclovir Synergistic Anemia, neutropenia 3
myelosuppression
Omeprazole 2 MMF drug exposure 2 efficacy 3
1, Avoid combination; 2, usually avoid (use only no other alternative agents available); 3, monitor closely; 4, no action needed (the risk for adverse drug reaction is
small). ACEI, angiotensin-converting enzyme inhibitor; AZA, azathioprine.
248 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

prednisone, and polymorphism of the UDP glucurono- Common Drug–Drug Interactions


syltransferase enzymatic system may significantly CsA, tacrolimus, and sirolimus are most com-
change the free fraction concentration of mycopheno- monly used with one or two other agents (azathio-
late. In addition, the total exposure to mycophenolate prine, MMF, sirolimus, or corticosteroids) to prevent
(AUC) does not seem to correlate to trough plasma acute rejection. The significant reduction of acute
concentration. Finally, neither the incidence of acute allograft rejection and graft loss from immunologic
rejection nor major adverse effects of mycophenolate factors has been paralleled with an increase in predomi-
seem to be related to plasma concentration. These nantly cardiovascular complications (hypertension, hy-
elements make therapeutic drug monitoring of myco- percholesterolemia, obesity, and posttransplantation dia-
phenolate unreliable and most likely would not opti- betes) and osteoporosis in kidney transplant recipients.
mize mycophenolate therapy or decrease drug toxici- Because most transplant patients have several comorbid
ties. conditions, avoidance of drug interactions is vital and
promotes allograft and patient survival. As the number
TOR Inhibitors: Sirolimus and Everolimus
Sirolimus and everolimus are novel macrolide of drugs for the management of comorbid conditions
immunosuppressants developed for use in combina- in kidney transplant recipients increases, so does the
tion with other immunosuppressive agents for preven- risk for clinically relevant drug interactions. The ma-
tion of acute rejection and slowing the progression of jority of immunosuppressive drugs are substrates for
chronic allograft nephropathy in kidney transplant commonly used drugs for the management of hyper-
recipients. At the molecular and cellular levels, these tension, hyperlipidemia, and infections (69).
agents inhibit mammalian TOR (mTOR), which is an The majority of clinically important immuno-
important protein for T cell proliferation. Initially, suppressive drug interactions results from induction
both sirolimus and everolimus bind to key regulatory or inhibition of immunosuppressive drug metabo-
enzyme of FK-BP-12; this complex then binds to lism; however, other mechanisms, including phar-
mTOR, which leads to cell-cycle arrest at the G1 macodynamic interactions, in which one agent may
stage. In addition to their immunosuppressive proper- alter the pharmacokinetic behavior of other drugs
ties, these agents seem to inhibit some of the vascular (absorption, distribution, metabolism, or elimina-
alterations of chronic allograft nephropathy. These tion) or have additive or synergistic toxicities, are
agents have no effect on the calcineurin phosphatase also important. To avoid drug– drug interactions,
pathway and are suitable agents to be used in combi- both pharmacokinetic properties and pharmacody-
nation with low-dosage CNIs or in calcineurin avoid- namic interactions should be considered (70). For
ance protocols (61). reduction of the severity of drug– drug interactions
Monitoring of sirolimus and everolimus trough in transplant recipients, it is crucial to identify the
blood levels has been helpful to adjust CNIs to target site and type of drug elimination with other con-
a lower plasma concentration. Sirolimus blood trough comitant immunosuppressant medications. Age-re-
levels should be maintained between 5 and 20 ng/ml, lated pharmacokinetics alterations, organ dysfunc-
and everolimus blood trough concentration should be tion, and comorbidities may also increase the risk
maintained between 3 and 8 ng/ml, depending on for potential drug interactions. A basic understand-
institution-specific protocols (62– 65). ing of pharmacokinetic and pharmacodynamic drug
The most common adverse events associated interaction may improve patient outcomes and de-
with use of sirolimus include myelosuppression, pri- crease the risk for drug toxicities. The most impor-
marily thrombocytopenia, hyperlipidemia, and de- tant principal of understanding drug interactions is
layed wound healing. Moderate to severe sirolimus to determine whether drug interactions are clinically
toxicity is characterized by interstitial pneumonitis, relevant and should be avoided. Although drug
Pneumocystis jiroveci pneumonia, and fungal infec- interaction programs or handheld devices and appli-
tions. Despite some clinical benefits, the use of siroli- cations provide an alert for drug interactions, they
mus as primary immunosuppression should be re- often do not provide any guidelines for management
served for patients with complications of standard CNI of drug interactions or take into account individual
protocols (66 – 68). patient factors (70) (Table 15).
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 249

Table 15. Sirolimus drug– drug interactions


Drug Mechanism Effects Severity Comments
ACEIs Synergistic Anemia, neutropenia 3 1 bone marrow
myelosuppression toxicity
Amprenavir 1 plasma level of sirolimus Hyperlipidemia, anemia, 3
neutropenia
Bromocriptine 1 plasma level of sirolimus Hyperlipidemia, anemia, 3
neutropenia
Carbamazepine 2 intestinal absorption 2 sirolimus level 2
Cholestyramine 2 intestinal absorption 2 sirolimus level 3
Clarithromycin 1 plasma level of sirolimus Hyperlipidemia, anemia, 2 Azithromycin is
neutropenia preferred agent
Co-trimoxazole Synergistic Anemia, neutropenia 3
myelosuppression
CsA 1 plasma level of sirolimus Hyperlipidemia, anemia, 3 Give 4 hours
neutropenia after the dose
Danazol 2 intestinal absorption 2 sirolimus level 3
Diltiazem 1 plasma level of sirolimus Hyperlipidemia, anemia, 2 Amlodipine is the
neutropenia preferred agent
Erythromycin 1 plasma level of sirolimus Hyperlipidemia, anemia, 2 Azithromycin is
neutropenia preferred agent
Fluconazole 1 plasma level of sirolimus Hyperlipidemia, anemia, 2
neutropenia
Ganciclovir Synergistic Anemia, neutropenia 3
myelosuppression
Indinavir 1 plasma level of sirolimus Hyperlipidemia, anemia, 2
neutropenia
Itraconazole 1 plasma level of sirolimus Hyperlipidemia, anemia, 2
neutropenia
Metoclopramide 1 plasma level of sirolimus Hyperlipidemia, anemia, 3
neutropenia
Nicardipine 1 plasma level of sirolimus Hyperlipidemia, anemia, 2 Amlodipine is
neutropenia preferred agent
Phenobarbital 1 metabolism of sirolimus 2 sirolimus level 2
Phenytoin 1 metabolism of sirolimus 2 sirolimus level 2
Rifabutin 1 metabolism of sirolimus 2 sirolimus level 2
Rifampin 1 metabolism of sirolimus 2 sirolimus level 2
Ritonavir 1 plasma level of sirolimus Hyperlipidemia, 2
Anemia, neutropenia
Verapamil 1 plasma level of sirolimus Hyperlipidemia, 2
Anemia, neutropenia
Voriconazole 1 plasma level of sirolimus Hyperlipidemia, anemia, 2
neutropenia
1, Avoid combination; 2, usually avoid (use only no other alternative agents available); 3, monitor closely; 4, no action needed (the risk for adverse drug reaction is
small). ACEI, angiotensin-converting enzyme inhibitor.

Pharmacokinetic Drug Interactions most therapeutic agents, patients who take immuno-
Many drugs may stimulate or inhibit the activity suppressive drugs may metabolize these drugs at a
of a variety of cytochrome P450 (CYP) enzymes, faster or slower rate and would be at risk for acute
including CYP3A4, CYP2C9, and CYP2C19. More rejection, allograft loss, and/or drug toxicity. Variabil-
than 50% of all drugs are metabolized through cyto- ity in elimination and drug– drug interactions has been
chrome P450 enzymes. Because these enzymes are observed in metabolism of a drug as a result of
important in the biotransformation and metabolism of polymorphism of cytochrome P450 expression, and
250 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

polymorphisms of cytochrome P isoenzyme and P- ceive co-administration of statins, azole antifungals,


glycoproteins may directly and/or indirectly influence and CsA should be closely monitored (75).
drug– drug interactions in transplant patients (71,72). Finally, St. John’s Wort can increase metabo-
Immunosuppressants may also stimulate or in- lism of CsA and tacrolimus by inducing both CYP-
hibit metabolism or prevent enterohepatic recircula- 450 IIIA and P-glycoprotein, whereas grapefruit can
tion of other immunosuppressant agents (Table 16). decrease metabolism of CsA and tacrolimus by
For example, corticosteroids at low dosages may in- inhibiting both CYP-450 IIIA and P-glycoprotein.
duce uridine diphosphate– glucuronosyltransferase en- Transplant patients should routinely be asked about use
zymes. Patients who are on steroid-free protocols have of herbal supplements and other supplements in addition
a higher incidence of myelosuppression and bone to other drugs (76).
marrow toxicities as a result of higher mycophenolate In addition to the numerous pharmacokinetic
plasma concentrations (73). interactions seen with maintenance immunosuppres-
Although there is no clinically important drug sants, there is the possibility for pharmacodynamic
interaction between mycophenolate and tacrolimus, interactions. Pharmacodynamic drug interactions
CsA may inhibit recirculation of mycophenolate and are common and predictable and for minimization
reduce MPA plasma concentrations (74). As a conse- of additive toxicities should be avoided completely
quence, stable patients who are on mycophenolate and if possible; if not, then toxicity should be monitored
CsA may experience adverse drug reactions from very closely. The use of other nephrotoxic and/or
mycophenolate after switching from CsA to tacrolimus. myelosuppressive agents should be used with cau-
A list of common drug interactions between immunosup- tion. For example, amphotericin B, aminoglyco-
pressive drugs are reviewed in Table 16 (73). sides, foscarnet, cidofovir, and nonsteroidal anti-
CsA may augment plasma hepatic hydroxy- inflammatory drugs may potentiate the neph-
methyl glutaryl–CoA reductase inhibitor (statin) con- rotoxicity of CNIs and should be avoided unless it is
centrations and increase the risk for myopathy and necessary for life-threatening situations. Use of gan-
rhabdomyolysis. Tacrolimus has a very limited effect ciclovir, valganciclovir, and co-trimoxazole in com-
on metabolism of statins, and switching patients from bination with mycophenolate or azathioprine can
tacrolimus to CsA can lead to increased risk for lead to an increased risk for myelosuppression in
statin-associated rhabdomyolysis. Patients who re- transplant patients (69,70).

Table 16. Pharmacokinetic drug interaction between immunosuppressive agents


Drug Interactions Effect Mechanism
CsA Mycophenolate 2 MPA exposure Inhibition of enterohepatic circulation
of MPA/MPAG
CsA Sirolimus 1 sirolimus exposure 2 metabolism of sirolimus through
CYP-450/P-gp
CsA Corticosteroids 2 CsA exposure 1 metabolism of CSA through CYP-
450/P-gp
CsA High-dosage corticosteroids 1 CsA exposure 2 metabolism of CSA through CYP-
for rejection 450/P-gp
Tacrolimus Mycophenolate 7 No drug interactions
Tacrolimus Sirolimus 2 tacrolimus exposure 1 metabolism of tacrolimus through
CYP-450/P-gp
Tacrolimus Corticosteroids 2 tacrolimus exposure 1 metabolism of tacrolimus through
CYP-450/P-gp
Sirolimus Corticosteroids 1 prednisone exposure 2 metabolism of prednisone
Sirolimus Mycophenolate 7 No pharmacokinetic drug
interactionsa
Mycophenolate Corticosteroids 2 MPA exposure UGT induction, increase MPA level
CYP-450, cytochrome P 450; MPA, mycophenolic acid; MPAG, mycophenolate glucuronide; P-gp, P-glycoprotein; UGT, UDP glucuronosyltransferase.
a
Pharmacodynamic drug interactions; a higher rate of myelosuppression.
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 251

References thymoglobulin for induction therapy in pancreas transplantation.


1. Potter M: The early history of plasma cell tumors in mice, 1954 – Transplant Proc 33: 1910, 2001
1976. Adv Cancer Res 98: 17–51, 2007 22. Webster A, Pankhurst T, Rinaldi F, Chapman JR, Craig JC: Poly-
2. Olyaei AJ, de Mattos AM, Bennett WM: Immunosuppressant-in- clonal and monoclonal antibodies for treating acute rejection episodes
duced nephropathy: Pathophysiology, incidence and management. in kidney transplant recipients. Cochrane Database Syst Rev (2):
Drug Saf 21: 471– 488, 1999 CD004756, 2006
3. Burk ML, Matuszewski KA: Muromonab-CD3 and antithymocyte 23. Burk ML, Matuszewski KA: Muromonab-CD3 and antithymocyte
globulin in renal transplantation. Ann Pharmacother 31: 1370 –1377, globulin in renal transplantation. Ann Pharmacother 31: 1370 –1377,
1997 1997
4. Lopez-Larrea C, Ortega F: Advances in translational transplant im- 24. Parlevliet KJ, Schellekens PT: Monoclonal antibodies in renal trans-
munology. Transplantation 88: S1–S7, 2009 plantation: A review. Transpl Int 5: 234 –246, 1992
5. Aull MJ, Suthanthiran M: Transplantation: Personalizing induction 25. Pham PT, Lipshutz GS, Pham PT, Kawahji J, Singer JS, Pham PC:
therapy in kidney transplantation. Nat Rev Nephrol 5: 619 – 620, 2009 The evolving role of alemtuzumab (Campath-1H) in renal transplan-
6. Golconda MS, de Mattos AM, Prather J, Olyaei AJ, Fletcher L, Head tation. Drug Des Dev Ther 3: 41– 49, 2009
MA, Wetzsteon P, Barry JM, Norman DJ: Renal transplantation at 26. Munoz AS, Cabanayan-Casasola CB, Danguilan RA, Padua FB, Ona
Oregon Health and Science University: Recent results and protocols. ET: Campath-1H (alemtuzumab) as an induction agent for the pre-
Clin Transpl 149 –154, 2003 vention of graft rejection and preservation of renal function in kidney
7. Shou ZF, Zhou Q, Cai JR, Cheng J, He Q, Wu JY, Chen JH: Efficacy transplant patients: Philippine 3-year follow-up. Transplant Proc 40:
and safety of induction therapy with alemtuzumab in kidney trans- 2230 –2233, 2008
plantation: A meta-analysis. Chin Med J (Engl) 122: 1692–1698, 27. Ortiz J, Palma-Vargas J, Wright F, Bingaman A, Agha I, Rosenblatt
2009 S, Foster P: Campath induction for kidney transplantation: Report of
8. Koch M, Becker T, Lueck R, Neipp M, Klempnauer J, Nashan B: 297 cases. Transplantation 85: 1550 –1556, 2008
Basiliximab induction therapy in kidney transplantation: Benefits for 28. Margreiter R, Klempnauer J, Neuhaus P, Muehlbacher F, Boesmuel-
long term allograft function after 10 years? Biologics 3: 51–56, 2009 ler C, Calne RY: Alemtuzumab (Campath-1H) and tacrolimus mono-
9. Padiyar A, Augustine JJ, Hricik DE: Induction antibody therapy in therapy after renal transplantation: Results of a prospective random-
kidney transplantation. Am J Kidney Dis 54: 935–944, 2009 ized trial. Am J Transplant 8: 1480 –1485, 2008
10. Olyaei AJ, Thi K, de Mattos AM, Bennett WM: Use of basiliximab 29. Kahan BD: Forty years of publication of Transplantation Proceed-
and daclizumab in kidney transplantation. Prog Transplant 11: 33– ings: The second decade—The cyclosporine revolution. Transplant
37, 2001 Proc 41: 1423–1437, 2009
11. Pescovitz MD: Use of antibody induction in pediatric renal trans- 30. Michelakis AM, McAllister RG: The effect of chronic adrenergic
plantation. Curr Opin Organ Transplant 13: 495– 499, 2008 receptor blockade on plasma renin activity in man. J Clin Endocrinol
12. Sandrini S: Use of IL-2 receptor antagonists to reduce delayed graft Metabol 34: 386 –394, 1972
function following renal transplantation: A review. Clin Transplant 31. Kuypers DR: Immunosuppressive drug monitoring: What to use in
19: 705–710, 2005 clinical practice today to improve renal graft outcome. Transpl Int 18:
13. Van Gelder T, Warle M, Ter Meulen RG: Anti-interleukin-2 receptor 140 –150, 2005
antibodies in transplantation: What is the basis for choice? Drugs 64: 32. Nashan B, Bock A, Bosmans JL, Budde K, Fijter H, Jaques B,
1737–1741, 2004 Johnston A, Lück R, Midtvedt K, Pallardó LM, Ready A, Salamé E,
14. Cibrik DM, Kaplan B, Meier-Kriesche HU: Role of anti-interleukin-2 Salizzoni M, Suarez F, Thervet E: Use of Neoral C monitoring: A
receptor antibodies in kidney transplantation. BioDrugs 15: 655– 666, European consensus. Transpl Int 18: 768 –778, 2005
2001 33. Billaud EM: C2 versus C0 cyclosporine monitoring: Still not the end.
15. Noel C, Abramowicz D, Durand D, Mourad G, Lang P, Kessler M, Transplantation 80: 542–544, 2005
Charpentier B, Touchard G, Berthoux F, Merville P, Ouali N, 34. Ponticelli C: Cyclosporine: From renal transplantation to autoim-
Squifflet JP, Bayle F, Wissing KM, Hazzan M: Daclizumab versus mune diseases. Ann N Y Acad Sci 1051: 551–558, 2005
antithymocyte globulin in high-immunological-risk renal transplant 35. Schiff J, Cole E, Cantarovich M: Therapeutic monitoring of cal-
recipients. J Am Soc Nephrol 20: 1385–1392, 2009 cineurin inhibitors for the nephrologist. Clin J Am Soc Nephrol 2:
16. Vincenti F, Kirkman R, Light S, Bumgardner G, Pescovitz M, 374 –384, 2007
Halloran P, Neylan J, Wilkinson A, Ekberg H, Gaston R, Backman L, 36. Naesens M, Kuypers DR, Sarwal M: Calcineurin inhibitor nephro-
Burdick J: Interleukin-2-receptor blockade with daclizumab to pre- toxicity. Clin J Am Soc Nephrol 4: 481–508, 2009
vent acute rejection in renal transplantation. Daclizumab Triple 37. Williams D, Haragsim L: Calcineurin nephrotoxicity. Adv Chronic
Therapy Study Group. N Engl J Med 338: 161–165, 1998 Kidney Dis 13: 47–55, 2006
17. Bumgardner GL, Hardie I, Johnson RW, Lin A, Nashan B, Pescovitz 38. Baluja P, Haragsim L, Laszik Z: Chronic allograft nephropathy. Adv
MD, Ramos E, Vincenti F, Phase III Daclizumab Study Group: Chronic Kidney Dis 13: 56 – 61, 2006
Results of 3-year phase III clinical trials with daclizumab prophylaxis 39. Morales JM, Dominguez-Gil B: Cardiovascular risk profile with the
for prevention of acute rejection after renal transplantation. Trans- new immunosuppressive combinations after renal transplantation.
plantation 72: 839 – 845, 2001 J Hypertens 23: 1609 –1616, 2005
18. Lin M, Ming A, Zhao M: Two-dose basiliximab compared with 40. Olyaei AJ, de Mattos AM, Bennett WM: Nephrotoxicity of immu-
two-dose daclizumab in renal transplantation: A clinical study. Clin nosuppressive drugs: New insight and preventive strategies. Curr
Transplant 20: 325–329, 2006 Opin Crit Care 7: 384 –389, 2001
19. Deeks ED, Keating GM: Rabbit antithymocyte globulin (thymo- 41. Christians U, Schmitz V, Schoning W, Bendrick-Peart J, Klawitter J,
globulin): A review of its use in the prevention and treatment of acute Haschke M, Klawitter J: Toxicodynamic therapeutic drug monitoring
renal allograft rejection. Drugs 69: 1483–1512, 2009 of immunosuppressants: Promises, reality, and challenges. Ther Drug
20. Hardinger KL: Rabbit antithymocyte globulin induction therapy in Monit 30: 151–158, 2008
adult renal transplantation. Pharmacotherapy 26: 1771–1783, 2006 42. Kino T, Hatanaka H, Miyata S, Inamura N, Nishiyama M, Yajima T,
21. Wiland AM, Fink JC, Philosophe B, Farney AC, Schweitzer EJ, Goto T, Okuhara M, Kohsaka M, Aoki H, et al.: FK-506, a novel
Colonna JO, Weir MR, Bartlett ST: Peripheral administration of immunosuppressant isolated from a Streptomyces: II. Immunosup-
252 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

pressive effect of FK-506 in vitro. J Antibiot (Tokyo) 40: 1256 –1265, renal function in sirolimus-treated renal transplant patients after early
1987 cyclosporine elimination. Transplantation 74: 1560 –1567, 2002
43. Kino T, Hatanaka H, Hashimoto M, Nishiyama M, Goto T, Okuhara 63. Oberbauer R, Segoloni G, Campistol JM, Kreis H, Mota A, Lawen J,
M, Kohsaka M, Aoki H, Imanaka H: FK-506, a novel immunosup- Russ G, Grinyó JM, Stallone G, Hartmann A, Pinto JR, Chapman J,
pressant isolated from a Streptomyces: I. Fermentation, isolation, and Burke JT, Brault Y, Neylan JF, Rapamune Maintenance Regimen
physico-chemical and biological characteristics. J Antibiot (Tokyo) Study Group: Early cyclosporine withdrawal from a sirolimus-based
40: 1249 –1255, 1987 regimen results in better renal allograft survival and renal function at
44. Vicari-Christensen M, Repper S, Basile S, Young D: Tacrolimus: 48 months after transplantation. Transpl Int 18: 22–28, 2005
Review of pharmacokinetics, pharmacodynamics, and pharmacoge- 64. Schena FP, Pascoe MD, Alberu J, del Carmen RM, Oberbauer R,
netics to facilitate practitioners’ understanding and offer strategies for Brennan DC, Campistol JM, Racusen L, Polinsky MS, Goldberg-
educating patients and promoting adherence. Prog Transplant 19: Alberts R, Li H, Scarola J, Neylan JF, Sirolimus CONVERT Trial
277–284, 2009 Study Group: Conversion from calcineurin inhibitors to sirolimus
45. Bowman LJ, Brennan DC: The role of tacrolimus in renal transplan- maintenance therapy in renal allograft recipients: 24-Month efficacy
tation. Expert Opin Pharmacother 9: 635– 643, 2008 and safety results from the CONVERT trial. Transplantation 87:
46. Laskow DA, Vincenti F, Neylan JF, Mendez R, Matas AJ: An 233–242, 2009
open-label, concentration-ranging trial of FK506 in primary kidney 65. Pascual J: Everolimus in clinical practice–renal transplantation.
transplantation: A report of the United States Multicenter FK506 Nephrol Dial Transplant 21[Suppl 3]: iii18 –iii23, 2006
Kidney Transplant Group. Transplantation 62: 900 –905, 1996 66. Neff RT, Jindal RM, Yoo DY, Hurst FP, Agodoa LY, Abbott KC:
47. Vincenti F, Laskow DA, Neylan JF, Mendez R, Matas AJ: One-year Analysis of USRDS: Incidence and risk factors for Pneumocystis
follow-up of an open-label trial of FK506 for primary kidney trans- jiroveci pneumonia. Transplantation 88: 135–141, 2009
plantation: A report of the U.S. Multicenter FK506 Kidney Trans- 67. Otton J, Hayward CS, Keogh AM, Glanville AR, Macdonald PS:
plant Group. Transplantation 61: 1576 –1581, 1996 Everolimus-associated pneumonitis in 3 heart transplant recipients.
48. Pirsch JD, Miller J, Deierhoi MH, Vincenti F, Filo RS: A comparison J Heart Lung Transplant 28: 104 –106, 2009
of tacrolimus (FK506) and cyclosporine for immunosuppression after 68. Flechner SM: Risk factors for and management of sirolimus-associ-
cadaveric renal transplantation. FK506 Kidney Transplant Study ated pneumonitis in kidney transplant recipients. Nat Clin Pract
Nephrol 4: 250 –251, 2008
Group. Transplantation 63: 977–983, 1997
69. Kuypers DR: Immunotherapy in elderly transplant recipients: A
49. Behrendt KE: The Hatch-Waxman Act: Balancing competing inter-
guide to clinically significant drug interactions. Drugs Aging 26:
ests or survival of the fittest? Food Drug Law J 57: 247–271, 2002
715–737, 2009
50. Benson MD: Therapeutic equivalents in clinical practice. Int J Fertil
70. Manitpisitkul W, McCann E, Lee S, Weir MR: Drug interactions in
Womens Med 46: 89 –94, 2001
transplant patients: What everyone should know. Curr Opin Nephrol
51. Olyaei AJ, de Mattos AM, Bennett WM: The impact of generic drugs
Hypertens 18: 404 – 411, 2009
on the cost of transplantation medical care. Transplant Proc 31:
71. Wang J: CYP3A polymorphisms and immunosuppressive drugs in
31S–34S, 1999
solid-organ transplantation. Expert Rev Mol Diagn 9: 383–390, 2009
52. Bennett WM, Olyaei AJ: Pharmacoeconomics of immunosuppressive
72. Burckart GJ: Pharmacogenomics: The key to improved drug therapy
agents in renal transplant recipients. Transplant Proc 31: 6S, 1999
in transplant patients. Clin Lab Med 28: 411– 422, vi, 2008
53. Peters JR, Hixon DR, Conner DP, Davit BM, Catterson DM, Parise
73. Kuypers DR: Influence of interactions between immunosuppressive
CM: Generic drugs: Safe, effective, and affordable. Dermatol Ther
drugs on therapeutic drug monitoring. Ann Transplant 13: 11–18,
22: 229 –240, 2009
2008
54. Banahan BF 3rd, Kolassa EM: A physician survey on generic drugs 74. Van Gelder T, Klupp J, Barten MJ, Christians U, Morris RE:
and substitution of critical dose medications. Arch Intern Med 157: Comparison of the effects of tacrolimus and cyclosporine on the
2080 –2088, 1997 pharmacokinetics of mycophenolic acid. Ther Drug Monit 23: 119 –
55. Davit BM, Nwakama PE, Buehler GJ, Conner DP, Haidar SH, Patel 128, 2001
DT, Yang Y, Yu LX, Woodcock J: Comparing generic and innovator 75. Kanbay M, Turgut F, Covic A, Goldsmith D: Statin treatment for
drugs: A review of 12 years of bioequivalence data from the United dyslipidemia in chronic kidney disease and renal transplantation: A
States Food and Drug Administration. Ann Pharmacother 43: 1583– review of the evidence. J Nephrol 22: 598 – 609, 2009
1597, 2009 76. Nowack R: Review article: Cytochrome P450 enzyme, and transport
56. Olyaei AJ, de Mattos AM, Bennett WM: Pharmacology of immuno- protein mediated herb– drug interactions in renal transplant patients:
suppressive drugs. Drugs Today (Barc) 34: 463– 479, 1998 Grapefruit juice, St. John’s Wort—and beyond! Nephrology (Carl-
57. de Mattos AM, Olyaei AJ, Bennett WM: Pharmacology of immuno- ton) 13: 337–347, 2008
suppressive medications used in renal diseases and transplantation.
Am J Kidney Dis 28: 631– 667, 1996
58. Levidiotis V, Chang S, McDonald S: Pregnancy and maternal out- Cardiovascular Medications
comes among kidney transplant recipients. J Am Soc Nephrol 20:
2433–2440, 2009 Antihypertensive Agents
59. Bunnapradist S, Lentine KL, Burroughs TE, Pinsky BW, Hardinger
KL, Brennan DC, Schnitzler MA: Mycophenolate mofetil dose re- Peripheral ␣ Blockers
ductions and discontinuations after gastrointestinal complications are The peripheral ␣ blockers prazosin, terazosin,
associated with renal transplant graft failure. Transplantation 82: and doxazosin undergo extensive hepatic metabolism
102–107, 2006
60. Van Gelder T: Mycophenolate blood level monitoring: Recent with pharmacokinetics that are not altered by the
progress. Am J Transplant 9: 1495–1499, 2009 presence of renal insufficiency; thus, peripheral ␣
61. Cowan PA, Heizer KE: Sirolimus: Mammalian target of rapamycin blockers do not require dosage adjustment in patients
inhibitor to prevent kidney rejection. Nephrol Nurs J 27: 623– 625,
2000 with reduced GFR. These drugs are also not apprecia-
62. Gonwa TA, Hricik DE, Brinker K, Grinyo JM, Schena FP: Improved bly dialyzed. They are used with some regularity in the
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 253

patient with hypertension and chronic kidney disease able in the patient with CKD unless combined with a
(CKD) in that they are useful add-on compounds for diuretic (7,8). The selection of a ␤ blocker for a patient
treatment-resistant hypertension. Use of ␣-adrenergic with CKD should occur with some knowledge of the
antagonists in the treatment of hypertension has been elimination characteristics of the drug as well as
limited by their tendency to increase plasma volume, a whether the compound has active metabolites (Table 17)
phenomenon that may be more evident at high dosages (9). Accumulation of a renally cleared ␤ blocker, such
and in patients with CKD (1). as atenolol, in a patient with CKD does not typically
improve BP control but can be associated with more
Central ␣ Agonists
frequent concentration-dependent adverse effects (9).
Central ␣ agonists, such as clonidine and guan-
In general, there is not a best ␤ blocker to use for the
facine, are used in the management of hypertension in
patient with CKD even as there is a growing appreci-
patients with CKD. Clonidine is available as a trans-
ation of the pharmacokinetic and pharmacodynamic
dermal patch, which is best absorbed from a chest or
heterogeneity for drugs in this class.
upper arm location. Transdermal clonidine is of par-
ticular utility for the treatment of the labile patient who Calcium Channel Blockers
has hypertension and requires multiple medications, Calcium channel blockers (CCBs) are commonly
the hospitalized patient who cannot take oral medica- used drugs in the patient with CKD, in part as a result
tions, the patient with prominent early morning BP of the predictability of their BP-lowering response.
surges, and/or the patient with erratic medication in- Also, coronary artery disease is common in the patient
take (2). It is also of particular utility when it is with CKD, and drugs in this class are effective anti-
necessary to supervise medication intake as can occur anginal agents. In general, the volume of distribution
in the nonadherent hemodialysis patient (3). (Vd), protein binding, and plasma half-life of CCBs
Guanfacine is predominantly hepatically cleared are comparable in patients with CKD and those with
and does not accumulate in CKD. Clonidine, unlike normal renal function with a few notable exceptions
guanfacine, undergoes modest renal clearance, and its that do not mandate dosage adjustment on the basis of
plasma half-life is somewhat prolonged in CKD, al- pharmacokinetic considerations (Table 18) (10). One
though there are no specific recommendations for
dosage adjustment in this population (4). Patients who
have CKD and suddenly stop oral clonidine can be Table 17. Elimination characteristics of ␤ blockers
expected to have less frequent rebound hypertension Active Accumulation in
relating to this delayed clearance compared with indi- Drug Metabolites Renal Disease
viduals with normal kidney function; however, this Acebutolol Yes Yes
has not been formally studied. Atenolol No Yes
Clonidine is typically dosed to BP effect and can Betaxolol No Yes
be expected to accumulate in the patient with CKD. Its Bisoprolol No Yes
use in patients with CKD seems not to be associated Carteolol Yes Yes
with so-called “paradoxical hypertension,” a phenom- Carvedilol Yes No
enon that occurs at very high plasma clonidine levels Celiprolol Yes No
Esmolol No No
related to there being sufficient peripheral ␣-receptor
Labetalol No No
stimulation such that the benefits derived from central Metoprolol No No
␣1- and ␣2-adrenergic receptors stimulation are over- Metoprolol LA No No
whelmed (5). Patients with CKD and sinus node dys- Nadolol No Yes
function are at risk for significant bradycardia with Nebivolol No No
clonidine, so its use in such patients is best avoided (6). Oxprenolol No No
Penbutolol No No
␤ Blockers Pindolol No No
␤ Blockers are commonly used drugs in the Propranolol Yes No
patient with CKD for the treatment of hypertension as Propranolol LA Yes No
well as for their cardioprotective effects. The BP- Sotalol No Yes
lowering effect of ␤ blockers is somewhat unpredict- Timolol No No
254 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

Table 18. Elimination characteristics of CCBs


Renal Failure
Normal Half-life Half-life Dosage Adjustment in
Drug Trade Name (hours)a (hours) Renal Failure
Amlodipineb Norvasc 40.0 to 50.0 50.0 No
Diltiazemc Cardizem 2.0 to 5.0 2.0 to 4.0 No
Felodipine Plendil 11.0 to 16.0 18.0 ⫾ 11.4 No
Isradipine Dynacirc 8.0 3.1 No
Nicardipine Cardene 11.5 Not available No; careful dosage titration
recommended
Nifedipine Procardia 2.0 4.0 No
Nimodipine Nimotop 2.8 22.0 No; close monitoring for BP
effect advisable
Nisoldipine Sular 15.0 Not available No
Nitrendipine 3.6 4.1 No
Verapamild Calan, Isoptin, Verelan, 3.0 to 7.0 (acute) 11.4 ⫾ 4.0 (15.2) No
Covera 8.0 to 12.0 (chronic)
a
Normal half-life values are taken from populations studied simultaneously with patients with renal failure or from comparative studies in the literature.
b
All data are from individuals with normal renal function other than for renal failure half-life.
c
Diltiazem has an active metabolite, desacetyldiltiazem.
d
Verapamil and norverapamil half-life values are reported. Acute and chronic dosing half-life values are reported in individuals with normal renal function.

exception is nicardipine, for which CYP3A4 metabo- sively renal with varying degrees of filtration and
lism and thereby plasma clearance are decreased in tubular secretion. There are two ACEIs with dual route
patients with CKD compared with normal individuals. of elimination, fosinopril and trandolapril, whose ac-
Although the mechanism of this defect in drug clear- tive diacid form is both hepatically and renally cleared
ance remains unclear, because enzyme activity has not (13,14). This property of combined renal and hepatic
been specifically studied, this defect in plasma clear- elimination minimizes accumulation in CKD once
ance is corrected by hemodialysis, suggesting the dosing to steady state has occurred. To date, no spe-
presence of a dialyzable inhibitor of nicardipine clear- cific adverse effect has been identified from ACEI
ance in advanced renal failure (11). accumulation, although cough has been suggested but
CCB-related adverse effects have to be consid- not proved to be an ACEI concentration– dependent
ered when these drugs are used in the patient with adverse effect. Angioneurotic edema is a potentially
CKD. Many patients with CKD tend to be constipated, life-threatening adverse effect with ACEIs that is more
and this can be aggravated by verapamil. Also, CCBs independent of ACEI dosage and is more common in
can produce peripheral edema, which sometimes pre- black patients (15). It is probable, though, that the
sents an interpretive problem in the typically already longer drug concentrations remain elevated once a
volume-expanded patient with CKD. response to an ACEI has occurred, the more likely it is
Angiotensin-Converting Enzyme Inhibitors that BP, renal function, and potassium handling will be
Angiotensin-converting enzyme inhibitors (ACEIs) negatively affected. Some patients are very sensitive
are frequently administered to patients with CKD for to the effects of an ACEI, particularly those who have
treatment of hypertension and their cardiorenal protec- an activated renin-angiotensin-aldosterone system;
tive effects. The BP-lowering effect of ACEIs is gen- thus, even minimal degrees of ACEI accumulation can
erally less in volume-expanded forms of hypertension, present a problem (16).
as is often the case in CKD, compared with other The current product label recommendations,
forms of hypertension; however, addition of a diuretic which suggest that ACEI dosages should be reduced in
to an ACEI (or angiotensin receptor blocker [ARB]) moderate to severe CKD, vary somewhat from com-
typically improves the BP-lowering response (12). pound to compound (Table 19). These differing dos-
For most ACEIs, elimination is almost exclu- age recommendations are inconsequential to the clin-
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 255

ical use of ACEIs in patients with CKD, though,

Usual dosage titration to effect


Titrate to maximum of 40 mg

Titrate to maximum of 15 mg
Titration (mg) in Renal
Recommended Dosage
because ACEIs are typically titrated to effect. Whereas

Titrate to optimal response


Failure per Day parameters such as BP and renal function are sensitive
to the plasma concentration of an ACEI, hyperkalemia
may be less so (17). If hyperkalemia occurs with an
ACEI, then a reduced dosage or use of a nonaccumu-
lating ACEI can be considered.
Angiotensin Receptor Blockers
Like ACEIs, ARBs are generally less efficacious
in the treatment of hypertension in the presence of
CKD and oftentimes require addition of a diuretic to
maximize their BP-lowering effect (12). Most of these
(mg) in Renal Failure per Day (CrCl 0

drugs undergo significant hepatic elimination, with the


Usual Total Dosage and/or Range

2.0 (on dialysis day; CrCl ⬍15 ml/min)

exception of candesartan, telmisartan, and the E-3174


metabolite of losartan, which are 40, 60, and 50%
hepatically cleared, respectively. Irbesartan and telm-
to 10 ml/min)

isartan undergo the greatest degree of hepatic elimi-


nation among the ARBs, with each having ⬎95% of
their systemic clearance by hepatic elimination. Val-
50% of normal dose

sartan and eprosartan both are approximately 70%


No adjustment

cleared by the hepatic route (Table 20) (18). As with


ACEIs, the dosage of an ARB that is given to a patient
with CKD should be adjusted according to the level of
2.5 (1)
Same

Same
Same

Same
Same

effect, not on the basis of a predetermined plan to


maintain an arbitrary blood level (19). ARBs may
increase serum potassium values less than ACEIs in
patients with CKD (20).
3.75 (1) (CrCl ⬍40 ml/min)
(CrCl 10 to 30 ml/min)

2.0 (every other day) (CrCl


Renal Failure per Day

Direct Renin Inhibitors


and/or Range (mg) in

25% of normal dose (CrCl


75% of normal dose (CrCl
Usual Total Dosage

Aliskirin is the only compound currently mar-


keted in the United States in this class. This com-
10 to 50 ml/min)

15 to 29 ml/min)

pound is a highly specific in vitro inhibitor of both


⬍40 ml/min)
No adjustment

human and primate renin with an IC50 of 0.6


nmol/L. The capacity of this drug to reduce BP in
Table 19. Elimination characteristics of ACEIs

patients with hypertension is similar to (or slightly


2.5 (1)

0.5 (1)
5 (1)
5 (1)

better than) what is seen with standard therapeutic


dosages of various ACEIs and ARBs (21,22). Opin-
ions vary considerably on the utility of aliskiren,
ranging from enthusiasm for an antihypertensive
Prinivil, Zestril
Trade Name

agent with a “new” mechanism of action to a “so


what” attitude derived, in part, from its cost and
Monopril
Lotensin

Accupril
Capoten

Univasc
Vasotec

Aceon

Altace

Mavik

absence of outcomes data distinguishing it from


ACEIs or ARBs. There still remains the need to
CrCl, creatinine clearance.

understand better how this compound fits in the


management of hypertension and its effects on renal
Trandolapril

and cardiovascular outcomes. Aliskiren is not re-


Perindopril
Benazepril

Fosinopril

Moexipril
Lisinopril

Quinapril
Captopril

Enalapril

Ramipril

nally cleared; therefore, dosage adjustment is not


Drug

necessary for patients with CKD (23). The renal


effects of aliskiren seem similar to those of an ACEI
256 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

Table 20. Mode of elimination for ARBs


Drug Trade Name Renal Hepatic
Candesartan Atacand 60 40
Eprosartan Teveten 30 70 (unchanged)
Irbesartan Avapro 1 99 (2C9)
Losartan Cozaar 10 90 (2C9/3A4)
Olmesartan Benicar 35 to 50 50 to 65 (unchanged)
E-3174 Metabolite of losartan 50 50
Telmisartan Micardis 1 99 (unchanged)
Valsartan Diovan 30 70 (unchanged)

or an ARB. There seems to be an additive antipro- urea nitrogen values or measured GFR and most likely
teinuric effect with aliskiren and an ARB such as represent inhibition of tubular secretion of creatinine
losartan (24). by ranolazine or one of its metabolites (28). Ranola-
zine also modestly increases BP in patients with severe
Antianginal Agents renal insufficiency by an as-yet-undetermined mecha-
Patients with CKD have a high prevalence of
nism (28).
coronary artery disease (CAD) and tend to have more
severe CAD and coronary calcification as well as a Cholesterol-Lowering Agents
worse prognosis than do patients with other cardiovas- Abnormalities in cholesterol metabolism are
cular risk factors (25). Patients with CKD have gen- ubiquitous in practically all forms of renal disease
erally been excluded from major cardiovascular treat- (30). As such, treatment with statins is important and
ment trials; therefore, the interplay between the common in this patient population. Two subtypes of
pharmacokinetics and pharmacodynamics of various statins are currently on the US market: Fermentation
antianginal agents in the patient with CKD is typically derived, or natural, statins (lovastatin, pravastatin, and
inferred from data derived from patients with more simvastatin) and synthetic statins (atorvastatin, rosuv-
normal renal function (26). A number of antihyperten- astatin, and fluvastatin).
sive agents, particularly ␤ blockers and CCBs, are With the exception of pravastatin, all statins
used in the patient with symptomatic ischemic heart undergo extensive microsomal metabolism by CYP
disease (see the Antihypertensive Agents section). enzymes. Pravastatin is eliminated through both renal
Other antianginal agents of note include nitrates and and nonrenal routes, with renal clearance accounting
ranolazine, which are addressed herein. for 47% of its total systemic clearance (31). CYP3A4
The pharmacokinetics of isosorbide dinitrate and is an important enzyme in the metabolism of several of
its two active metabolites, isosorbide-2-nitrate and the hydroxymethyl glutaryl–CoA reductase inhibitors,
isosorbide-5-nitrate, are not altered by the presence of including atorvastatin, lovastatin, and simvastatin;
renal failure, and dosage adjustment is not required in therefore, these compounds are prone to drug– drug
such patients (27). Ranolazine is primarily metabo- interactions with CYP3A4 inhibitors, such as vera-
lized by CYP3A4 and is a substrate of P-glycoprotein. pamil and diltiazem (32,33).
In severe renal failure, there is upwards of a twofold Statins are generally well tolerated in patients
increase in the AUC for ranolazine; because ⬍7% of with CKD (34). The major adverse effect of statins is
an administered dose of ranolazine is excreted un- myopathy, often defined as muscle pain or weakness
changed, factors other than a reduced GFR contribute associated with creatine kinase levels 10 times higher
to the increase in ranolazine concentrations in renal than the upper limits of normal. Myotoxicity with
impairment (28). statins is a concentration-dependent phenomenon. Be-
Ranolazine has a very narrow therapeutic win- cause the symptoms that commonly are seen with
dow and should be dosed carefully in the patient with statin-related myopathy often are not different from
advanced CKD (28,29). Small increases in serum the musculoskeletal complaints that commonly are
creatinine values on the order of 0.1 to 0.2 mg/dl occur seen in the patient with CKD, there should be a low
with ranolazine without a concomitant change in blood threshold for measuring creatine kinase levels under
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 257

these conditions. In addition, the lower level of renal (37). These agents are associated with a reversible rise
function in these patients in and of itself makes it in serum creatinine (38,39); whether this it is due to a
important that myopathy be identified early, because real change in GFR is still debated. One proposed
the reduced renal function adversely affects the toler- explanation for this phenomenon is that fibrates in-
ance and risk for rhabdomyolysis (32). crease the production of creatinine, in which case a
Finally, the numbers of medicines that typically rise in serum creatinine values would not represent a
are prescribed to patients with renal failure increases true deterioration in renal function. An alternative
the chance for drug– drug interactions and the risk for theory is that fibrates reduce the production of vaso-
myopathy with statins. Adhering to current recom- dilatory prostaglandins, which could lead to a true
mendations for statin therapy in CKD should minimize change in renal function (39).
the risk for adverse reactions to statins in these patients Routine serum creatinine monitoring is advisable
(Table 21) Unfortunately, adherence to these dosage for fibrate-treated patients, particularly for those with
recommendations may limit achievement of choles- preexisting renal disease. A 30% increase in serum
terol goals; in fact, recent clinical trials have called creatinine values in the absence of other causes of
into question the routine use of statins in the patient
serum creatinine change warrants discontinuation of
with ESRD and may affect the overall use of these
fibrate therapy. Serum creatinine values can take sev-
compounds in such patients (35,36).
eral weeks to return to their baseline values after
Triglyceride-Lowering Agents discontinuation of a fibrate. There is little information
Fibrates are a class of lipid-lowering medication in the literature as to whether a patient who has
primarily used as second-line agents behind statins experienced a rise in serum creatinine with fibrate

Table 21. Dosage recommendations for statins in patients with normal renal function and with CKD/ESRD
Normal
Drug (mg/d) CKD/ESRD Transplantation
Atorvastatin 10 to 80 Dosage adjustment in patients with renal No comment
insufficiency is not necessary
Fluvastatin 20 to 80 Dosage adjustments for mild to No comment
moderate renal impairment are not
necessary; caution should be exercised
with severe impairment
Lovastatin 10 to 80 In patients with CrCl ⬍30 ml/min, In patients taking CsA, therapy should begin
dosage increases ⬎20 mg should be with 10 mg/d and should not exceed 20
carefully considered and if deemed mg/d
necessary should be supplemented
cautiously
Pravastatin 10 to 40 No comment In patients taking CsA, therapy should begin
with 10 mg and titration to higher dosages
should be done with caution
Rosuvastatin 5 to 40 For patients who have severe renal In patients taking CsA, dosage should be
impairment (CrCl ⬍30 ml/min per limited to 5 mg/d
1.73 m2) and are not on hemodialysis,
dosing should be started at 5 mg/d and
not exceed 10 mg/d
Simvastatin 5 to 80 Dosage adjustment should not be In patients taking CsA, therapy should begin
necessary in mild to moderate renal with 5 mg/d and not exceed 10 mg/d
insufficiency; in patients with severe
renal insufficiency, 5 mg/d should be
the starting dosage with close
monitoring if there is any titration
Recommendations taken directly from the package inserts for individual HMG-CoA reductase inhibitors. CrCl, creatinine clearance.
258 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

therapy can be safely restarted on a fibrate (39,40). If References


clinical circumstances dictate fibrate therapy, then gem- 1. Carlson RV, Bailey RR, Begg EJ, Cowlishaw MG, Sharman JR:
Pharmacokinetics and effect on blood pressure of doxazosin in
fibrozil would be the preferred agent in that this com- normal subjects and patients with renal failure. Clin Pharmacol Ther
pound has no recognizable effect on renal function (41). 40: 561–566, 1986
2. Mancia G, Parati G, Pomidossi G, Malaspina D, Colombo A,
Camesasca C, Groppelli A, Casadei R, Zanchetti A: Evaluation of the
Antiarrhythmic Agents antihypertensive effect of TTS clonidine by multiple 24-hour auto-
Both atrial and ventricular rhythm disturbances matic blood pressure monitoring. J Cardiovasc Pharmacol 10[Suppl
are common in patients with CKD, with as many as 13 12]: S187–S193, 1987
3. Sica DA, Grubbs R: Transdermal clonidine: therapeutic consider-
to 27% of patients who are on long-term hemodialysis ations. J Clin Hypertens (Greenwich) 7: 558 –562, 2005
having atrial fibrillation (42). The foremost safety 4. Lowenthal DT, Affrime MB, Meyer A, Kim KE, Falkner B, Sharif K:
issue with therapies that are used for atrial fibrillation Pharmacokinetics and pharmacodynamics of clonidine in varying
states of renal function. Chest 83[Suppl]: 386 –390, 1983
is the propensity of class IA and III agents to cause 5. Dire DJ, Kuhns DW: The use of sublingual nifedipine in a patient
torsade de pointes arrhythmias (43). The majority of with a clonidine overdose. J Emerg Med 6: 125–128, 1988
the compounds that are used in the treatment of atrial 6. Byrd BF 3rd, Collins HW, Primm RK: Risk factors for severe
bradycardia during oral clonidine therapy for hypertension. Arch
fibrillation have a narrow therapeutic window, and Intern Med 148: 729 –733, 1988
several of them are extensively renally cleared, includ- 7. Ritz E, Rump LC: Do beta-blockers combined with RAS inhibitors
ing procainamide and its N-acetylated metabolite, and make sense after all to protect against renal injury? Curr Hypertens
Rep 9: 409 – 414, 2007
newer agents such as dofetilide, flecainide, and D- 8. Bakris GL, Hart P, Ritz E: Beta blockers in the management of
sotalol. chronic kidney disease. Kidney Int 70: 1905–1913, 2006
As an example of the likelihood of proarrythmic 9. Frishman WH, Alwarshetty M: Beta-adrenergic blockers in systemic
hypertension: Pharmacokinetic considerations related to the current
events with class III agents in the setting of renal guidelines. Clin Pharmacokinet 41: 505–516, 2002
disease, dofetilide has the following black box warn- 10. Sica DA, Gehr TW: Calcium-channel blockers and end-stage renal
ing: “To minimize the risk of induced arrhythmia, disease: Pharmacokinetic and pharmacodynamic considerations.
Curr Opin Nephrol Hypertens 12: 123–131, 2003
patients initiated or re-initiated on dofetilide should be 11. Ahmed JH, Grant AC, Rodger RS, Murray GR, Elliott HL: Inhibitory
placed for a minimum of 3 days in a facility that can effect of uraemia on the hepatic clearance and metabolism of nicar-
provide calculations of creatinine clearance, continu- dipine. Br J Clin Pharmacol 32: 57– 62, 1991
12. Abe M, Okada K, Maruyama T, Matsumoto K: Renoprotect and
ous electrocardiographic monitoring, and cardiac re- blood pressure lowering effect of low-dose hydrochlorothiazide
suscitation.” Proarrhythmic effects of class III agents, added to intensive renin-angiotensin inhibition in hypertensive pa-
such as torsade de pointes, have been described as tients with chronic kidney disease. Int J Clin Pharmacol Ther 47:
525–532, 2009
being more common in certain patient types, including 13. Sica DA: Kinetics of angiotensin converting enzyme inhibitors in
women, and those with electrolyte disturbances such renal failure. J Cardiovasc Pharmacol 20[Supp 10]: S13–S20, 1992
as hypokalemia and hypomagnesemia, renal failure, 14. Sica DA, Cutler RE, Parmer RJ, Ford NF: Comparison of the steady
state pharmacokinetics of fosinopril, lisinopril and enalapril in pa-
and prolongation of the QT/QTc interval (44,45). tients with chronic renal insufficiency. Clin Pharmacokinet 20: 420 –
The best measurement tool for determining the 427, 1991
level of renal function for appropriate dosing of class 15. Gibbs CR, Lip GY, Beevers DG: Angioedema due to ACE inhibitors:
Increased risk in patients of African origin. Br J Clin Pharmacol 48:
III agents is not established. Reliance on creatinine 861– 865, 1999
clearance measurements, as is the suggestion for 16. Schoolwerth AC, Sica DA, Ballermann BJ, Wilcox CS: Renal con-
dofetilide, runs the risk for significantly overestimat- siderations in angiotensin converting enzyme inhibitor therapy: A
statement for healthcare professionals from the Council on the Kid-
ing the true GFR and dosing this compound too high; ney in Cardiovascular Disease and the Council for High Blood
accordingly, the use of electrocardiographic moni- Pressure Research of the American Heart Association. Circulation
toring becomes critical to improving the safety 104: 1985–1991, 2001
17. Bakris GL, Weir MR: Angiotensin-converting enzyme inhibitor-
profile of this compound in renal disease. These associated elevations in serum creatinine: Is this a cause for concern?
drugs probably should not be used in the advanced Arch Intern Med 160: 685– 693, 2000
stages of renal failure; dofetilide is contraindicated 18. Sica DA: Renal handling of angiotensin receptor blockers: Clinical
relevance. Curr Hypertens Rep 5: 337–339, 2003
in patients with a creatinine clearance of ⬍20 ml/ 19. Leidig M, Bambauer R, Kirchertz EJ, Szabã T, Handrock R, Leinung
min. Dialysis may be useful in the setting of renal D, Baier M, Schmieder RE: Efficacy, safety and tolerability of
failure and the development of torsade de pointes; valsartan 80 mg compared to irbesartan 150 mg in hypertensive
patients on long-term hemodialysis (VALID study). Clin Nephrol 69:
however, there is varying dialysance of the renally 425– 432, 2008
cleared class III agents (46). 20. Bakris GL, Siomos M, Richardson D, Janssen I, Bolton WK, Hebert
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 259

L, Agarwal R, Catanzaro D: ACE inhibition or angiotensin receptor mellitus (the FIELD study): Randomized controlled trial. Lancet 366:
blockade: Impact on potassium in renal failure. VAL-K Study Group. 1849 –1861, 2005
Kidney Int 58: 2084 –2092, 2000 39. Sica DA: Fibrate therapy and renal function. Curr Atheroscler Rep
21. Brown MJ: Aliskiren. Circulation 118: 773–784, 2008 11: 338 –342, 2009
22. Gradman AH, Kad R: Renin inhibition in hypertension. J Am Coll 40. Davidson MH, Armani A, McKenney JM, Jacobson TA: Safety
Cardiol 51: 519 –528, 2008 considerations with fibrate therapy. Am J Cardiol 99: 3C–18C, 2007
23. Vaidyanathan S, Bigler H, Yeh C, Bizot MN, Dieterich HA, Howard 41. Evans RJ, Forland SC, Cutler RE: The effect of renal function on the
D, Dole WP: Pharmacokinetics of the oral direct renin inhibitor pharmacokinetics of gemfibrozil. J Clin Pharmacol 27: 994 –1000,
aliskiren alone and in combination with irbesartan in renal impair- 1987
ment. Clin Pharmacokinet 46: 661– 675, 2007 42. Reinecke H, Brand E, Mesters R, Schäbitz WR, Fisher M, Pavenstädt
24. Parving HH, Persson F, Lewis JB, Lewis EJ, Hollenberg NK, H, Breithardt G: Dilemmas in the management of atrial fibrillation in
AVOID Study Investigators: Aliskiren combined with losartan in chronic kidney disease. J Am Soc Nephrol 20: 705–711, 2009
type 2 diabetes and nephropathy. N Engl J Med 358: 2433–2446, 43. Camm AJ: Safety considerations in the pharmacological management
2008 of atrial fibrillation. Int J Cardiol 127: 299 –306, 2008
25. Sarnak MJ, Levey AS, Schoolwerth AC, Coresh J, Culleton B, 44. Gupta A, Lawrence AT, Krishnan K, Kavinsky CJ, Trohman RG:
Hamm LL, McCullough PA, Kasiske BL, Kelepouris E, Klag MJ, Current concepts in the mechanisms and management of drug-
Parfrey P, Pfeffer M, Raij L, Spinosa DJ, Wilson PW: Kidney disease induced QT prolongation and torsade de pointes. Am Heart J 153:
as a risk factor for development of cardiovascular disease: A state- 891– 899, 2007
ment from the American Heart Association Councils on Kidney in 45. Dancey D, Wulffhart Z, McEwan P: Sotalol-induced torsades de
Cardiovascular Disease, High Blood Pressure Research, Clinical pointes in patients with renal failure. Can J Cardiol 13: 55–58, 1997
Cardiology, and Epidemiology and Prevention. Circulation 108: 46. van Uum SH, van den Merkhof LF, Lucassen AM, Wuis EW,
2154 –2169, 2003 Diemont W: Successful haemodialysis in sotalol-induced torsade de
26. Charytan D, Kuntz RE: The exclusion of patients with chronic kidney pointes in a patient with progressive renal failure. Nephrol Dial
disease from clinical trials in coronary artery disease. Kidney Int 70: Transplant 12: 331–333, 1997
2021–2030, 2006
27. Evers J, Bonn R, Boertz A, Cawello W, Dickmans HA, Weiss M:
Pharmacokinetics of isosorbide dinitrate, isosorbide-2-nitrate and Diuretics
isosorbide-5-nitrate in renal insufficiency after repeated oral dosage.
Klin Wochenschr 67: 342–348, 1989 Thiazide-Type Diuretics
28. Jerling M, Abdallah H: Effect of renal impairment on multiple-dose
Thiazide diuretics are not the diuretics of choice
pharmacokinetics of extended-release ranolazine. Clin Pharmacol
Ther 78: 288 –297, 2005 for patients with renal insufficiency, with the possible
29. Jerling M: Clinical pharmacokinetics of ranolazine. Clin Pharmaco- exception of the thiazide-like diuretics chlorthalidone
kinet 45: 469 – 491, 2006
and metolazone. This distinction among the thiazide-
30. Quaschning T, Krane V, Metzger T, Wanner C: Abnormalities in
uremic lipoprotein metabolism and its impact on cardiovascular type diuretics relates to the differing pharmacologic
disease. Am J Kid Dis 38[Suppl 1]: S14 –S19, 2001 characteristics of these compounds in that they have a
31. Halstenson CE, Triscari J, DeVault A, Shapiro B, Keane W, Pan H:
much larger volume of distribution and more extended
Single-dose pharmacokinetics of pravastatin and metabolites in pa-
tients with renal impairment. J Clin Pharmacol 32: 124 –132, 1992 delivery to their sites of action in the nephron (1).
32. Sica DA, Gehr TW: 3-Hydroxy-3-methylglutaryl coenzyme A reduc- Metolazone, which is often given together with a loop
tase inhibitors and rhabdomyolysis: Considerations in the renal fail- diuretic in diuretic-resistant states, is very poorly ab-
ure patient. Curr Opin Nephrol Hypertens 12: 123–133, 2002
33. Molden E, Skovlund E, Braathen P: Risk management of simvastatin sorbed from the gastrointestinal tract, which should be
or atorvastatin interactions with CYP3A4 inhibitors. Drug Saf 31: taken into account when determining the appropriate
587–596, 2008 dosage and frequency of dosing with this agent (2).
34. Navaneethan SD, Nigwekar SU, Perkovic V, Johnson DW, Craig JC,
Strippoli GF: HMG CoA reductase inhibitors (statins) for dialysis Although a large dose of a thiazide diuretic (50 to
patients. Cochrane Database Syst Rev (3): CD004289, 2009 100 mg) will initiate a diuresis in patients with mild
35. Krane V, Winkler K, Drechsler C, Lilienthal J, März W, Wanner C, renal insufficiency, the response in patients with a
German Diabetes and Dialysis Study Investigators: Effect of atorva-
statin on inflammation and outcome in patients with type 2 diabetes GFR of less than approximately 50 ml/min is generally
on hemodialysis. Kidney Int 74: 1461–1467, 2008 poor. Patients with chronic kidney disease (CKD) are
36. Fellström BC, Jardine AG, Schmieder RE, Holdaas H, Bannister K, not really “resistant“ to thiazide diuretics per se;
Beutler J, Chae DW, Chevaile A, Cobbe SM, Grönhagen-Riska C, De
Lima JJ, Lins R, Mayer G, McMahon AW, Parving HH, Remuzzi G, rather, the basis for failure of a thiazide diuretic is an
Samuelsson O, Sonkodi S, Sci D, Süleymanlar G, Tsakiris D, Tesar insufficient potency to meet the needs of patients with
V, Todorov V, Wiecek A, Wüthrich RP, Gottlow M, Johnsson E, a Na⫹-retaining state. Patients who receive fixed-
Zannad F, AURORA Study Group: Rosuvastatin and cardiovascular
events in patients undergoing hemodialysis. N Engl J Med 360: dosage combination antihypertensive therapy that con-
1395–1407, 2009 tains a thiazide diuretic should be considered for
37. Chapman MJ: Pharmacology of fenofibrate. Am J Med 83: 21–25, conversion to a loop diuretic (together with whatever
1987
38. The Field Study Investigators: Effects of long-term fenofibrate ther- was the other component of the fixed-dosage combi-
apy on cardiovascular events in 9795 people with type 2 diabetes nation) when the GFR drops below approximately 30
260 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

ml/min, particularly when BP control has been less problem is delivering adequate drug amounts of the
than desired (3). diuretic to the site of action.
A frequently posed question for a patient with
Loop Diuretics severe renal insufficiency is, “What is the largest
Loop diuretics are the most commonly used single dosage of a loop diuretic beyond which there is
diuretics in renal failure. Approximately 50% of a no additional yield?” The maximal natriuretic re-
dose of furosemide is excreted in the urine unchanged; sponse occurs with intravenous bolus dosages of 160
the remainder is renally conjugated to glucuronic acid. to 200 mg of furosemide or the equivalent dosages of
Therefore, in patients with renal failure, the plasma bumetanide (4 to 5 mg) and torsemide (50 to 100 mg),
half-life of furosemide is prolonged because both uri- and nothing is accomplished by using larger dosages.
nary excretion and renal conjugation are reduced. The Some patients may require dosages in the range of 160
two other loop diuretics that are available in the United to 200 mg several times a day to persist in their
States, bumetanide and torsemide, are largely hepati- diuresis (9). Single intravenous bolus dosages of 160
cally metabolized (50 and 80%, respectively), and to 200 mg can cause transient tinnitus, but this effect
their half-lives do not change appreciably in renal can be minimized by administering the dose over a
failure (4); however, renal insufficiency will impair period of 20 to 30 min at a rate no greater than 4
their tubular delivery. In addition to the routes of mg/min (10). Ototoxicity caused by ethacrynic acid
metabolism, the pharmacokinetic features of diuretics seems to develop more gradually and takes longer to
that assume clinical significance are bioavailability resolve than that caused by furosemide or bumetanide.
and half-life (5). The bioavailability of loop diuretics Limited data suggest that bumetanide may produce a
is not affected by renal insufficiency. lower incidence of ototoxicity. Finally, because poten-
On average, furosemide is 50% absorbed but tiation of the ototoxic effects of aminoglycosides and
within a range of 10 to 100% (6). This wide range loop diuretics is well documented, these drugs should
makes it a matter of some guesswork as to how much be co-administered cautiously (11).
furosemide will be absorbed in an individual patient,
and varied dosages of furosemide must be tried before Potassium-Sparing Diuretics
the drug is deemed ineffective. This issue is poten- Potassium-sparing diuretics are to be used cau-
tially further complicated by the presence of bowel tiously in patients with renal failure because of the risk
wall edema in the patient with decompensated heart for hyperkalemia. Amiloride is an organic cation that
failure, which may slow and diminish loop diuretic works as an antagonist of the epithelial sodium channel;
absorption from the gastrointestinal tract after oral it is both filtered and extensively secreted by the organic
administration (7). In contrast, absorption and bio- cation pathway. Renal disease prolongs its plasma half-
availability of bumetanide and torsemide are nearly life; accordingly, the dosage should be reduced by 50%
complete, ranging from 80 to 100%. in patients with a creatinine clearance of ⬍50 ml/min
Several pharmacodynamic features of diuretics (12). Amiloride can compete for tubular secretion with
are clinically important. In patients with a creatinine other organic cations, such as cimetidine, metformin, or
clearance of ⬍20 ml/min, delivery of a loop diuretic trimethoprim. Like all potassium-sparing diuretics, the
into the tubular fluid is only approximately 5 to 10% likelihood of its causing hyperkalemia is greatest in the
of what would be delivered in individuals with normal patient with CKD and diabetes (13).
GFR. Thus, a large dose must be given to attain a Triamterene, another potassium-sparing diuretic that
threshold quantity of diuretic in the tubular fluid for also inhibits the epithelial sodium channel, is seldom given
delivery to the diuretic site of action. The relation alone but rather is typically given together with hydrocholo-
between the rate at which the diuretic is excreted and rothiazide as a fixed-dosage combination product for the
the response in patients with renal insufficiency is treatment of hypertension. The pharmacokinetics of triam-
similar to what is observed in normal individuals (4,8). terene are complex, because it is hepatically converted to an
Thus, the remaining nephrons in patients with renal active metabolite, which then undergoes active tubular se-
insufficiency (unless there are other circumstances cretion. Renal disease impairs the tubular secretion of this
leading to sodium retention, such as nephrotic syn- metabolite, and lengthening of the dosage interval to every
drome) retain their responsiveness to diuretics; the 12 hours is suggested when the creatinine clearances is ⬍50
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 261

ml/min (14). Triamterene is also associated with crystalluria 3. Dussol B, Moussi-Frances J, Morange S, Somma-Delpero C, Mun-
dler O, Berland Y: A randomized trial of furosemide vs hydrochlo-
and occasionally with triamterene stones (15). A final con- rothiazide in patients with chronic renal failure and hypertension.
sideration with triamterene is its tendency to cause acute Nephrol Dial Transplant 20: 349 –353, 2005
renal failure when given together with a nonsteroidal anti- 4. Brater DC: Diuretic therapy. N Engl J Med 339: 387–395, 1998
5. Sica DA, Gehr TW: Diuretic combinations in refractory oedema
inflammatory drug relating to an intrinsic vasoconstrict- states: Pharmacokinetic-pharmacodynamic relationships. Clin Phar-
ing property that is unique to triamterene or its metabo- macokinet 30: 229 –249, 1996
lites (16). 6. Murray MD, Haag KM, Black PK, Hall SD, Brater DC: Variable
furosemide absorption and poor predictability of response in elderly
Spironolactone and the newer agent eplerenone differ patients. Pharmacotherapy 17: 98 –106, 1997
mechanistically from amiloride and triamterene in that they 7. Vasko MR, Cartwright DB, Knochel JP, Nixon JV, Brater DC:
are aldosterone receptor antagonists. This is the basis for Furosemide absorption altered in decompensated congestive heart
failure. Ann Intern Med 102: 314 –318, 1985
their expanding use in heart failure and therapy-resistant 8. Brater DC: Clinical pharmacology of loop diuretics in health and
hypertension (17). Whereas eplerenone is metabolized by disease. Eur Heart J 13[Suppl G]: 10 –14, 1992
CYP3A4 to inactive metabolites, spironolactone is oxida- 9. Shankar SS, Brater DC: Loop diuretics: From the Na-K-2Cl trans-
porter to clinical use. Am J Physiol Renal Physiol 284: F11–F21,
tively metabolized to two active metabolites, canrenone and 2003
7-␣-thiomethylspironolactone, with half-life values in the 10. Rybak LP: Ototoxicity of loop diuretics. Otolaryngol Clin North Am
26: 829 – 844, 1993
24-hour range. The pharmacokinetics of spironolactone and
11. Bates DE, Beaumont SJ, Baylis BW: Ototoxicity induced by genta-
eplerenone have not been studied in the setting of renal micin and furosemide. Ann Pharmacother 36: 446 – 451, 2002
failure. Dosage adjustment or use of spironolactone or 12. Spahn H, Reuter K, Mutschler E, Gerok W, Knauf H: Pharmacoki-
netics of amiloride in renal and hepatic disease. Eur J Clin Pharma-
eplerenone is not solely based on the level of renal function; col 33: 493– 498, 1987
rather, it is governed by the likelihood of clinically relevant 13. Greenberg A: Diuretic complications. Am J Med Sci 319: 10 –24,
hyperkalemia. Spironolactone can be given to patients with 2000
14. Sica DA, Gehr TW: Triamterene and the kidney. Nephron 51:
ESRD with minimal risk (18). Spironolactone and/or its 454 – 461, 1989
metabolites have a prolonged potassium-sparing effect that 15. Fairley KF, Woo KT, Birch DF, Leaker BR, Ratnaike S: Triam-
should be accounted for when it is prescribed. Eplerenone terene-induced crystalluria and cylinduria: Clinical and experimental
studies. Clin Nephrol 26: 169 –173, 1986
should be used carefully with inhibitors of CYP3A4, such as 16. Favre L, Glasson P, Vallotton MB: Reversible acute renal failure
verapamil and diltiazem, because these compounds would from combined triamterene and indomethacin: A study in healthy
be expected to inhibit the clearance of eplerenone and subjects. Ann Intern Med 96: 317–320, 1982
17. Nishizaka MK, Zaman MA, Calhoun DA: Efficacy of low-dose
therein extend its duration of action. spironolactone in subjects with resistant hypertension. Am J Hyper-
tens 16: 925–930, 2003
Mannitol 18. McLaughlin N, Gehr TW, Sica DA: Aldosterone-receptor antago-
nism and end-stage renal disease. Curr Hypertens Rep 6: 327–330,
Mannitol, an osmotic diuretic, exerts its diuretic 2004
effect at the proximal tubule and loop of Henle. If it 19. Borges HF, Hocks J, Kjellstrand CM: Mannitol intoxication in
goes unfiltered and instead remains in the circulation, patients with renal failure. Arch Intern Med 142: 63– 66, 1982
as in patients with reduced GFR, then it can increase
intravascular volume by an osmotic drag effect. The Gastrointestinal Agents
convective flux that occurs with high plasma mannitol Antacids are commonly used self-prescribed
concentrations will result in dilutional hyponatremia, medications. They consist of calcium carbonate and
increases in serum potassium, and, if concentrations magnesium and aluminum salts in various compounds
go high enough, acute renal failure, which occurs or combinations. Both magnesium- and aluminum-
secondary to afferent arteriolar vasoconstriction (19). containing antacids should be used sparingly, if at all,
The risks that are associated with mannitol, coupled in the patient with advanced-stage chronic kidney
with the availability of other highly effective diuretics, disease (CKD) (1). All antacids can produce drug
relegate its use to nondiuretic indications, such as interactions by changing gastric pH, thereby altering
cerebral edema. drug dissolution of dosage forms, decreasing acid
hydrolysis of drugs, or changing renal drug clearance
References by altering urinary pH. Most antacids, except sodium
1. Sica DA: Chlorthalidone: Has it always been the best thiazide-type bicarbonate, may decrease drug absorption by adsorp-
diuretic? Hypertension 47: 321–322, 2006
2. Sica DA: Metolazone and its role in edema management. Congest tion or chelation of other drugs. Antacid-related drug
Heart Fail 9: 100 –105, 2003 interactions, based on chelation of co-administered
262 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

medications, can be avoided by appropriately sched- 2. Reinke CM, Breitkreutz J, Leuenberger H: Aluminum in over-the-counter
drugs: Risks outweigh benefits? Drug Saf 26: 1011–1025, 2003
uling medication administration times. Aluminum ab- 3. Gladziwa U, Klotz U: Pharmacokinetics and pharmacodynamics of
sorption from antacids or other aluminum-containing H2-receptor antagonists in patients with renal insufficiency. Clin
compounds can be substantially enhanced by the con- Pharmacokinet 24: 319 –332, 1993
4. van Acker BA, Koomen GC, Koopman MG, de Waart DR, Arisz L:
comitant intake of citrate-containing products, so these Creatinine clearance during cimetidine administration for measure-
combinations must be avoided (2). ment of glomerular filtration rate. Lancet 340: 1326 –1329, 1992
Patients with CKD have a high prevalence of gas- 5. Härmark L, van der Wiel HE, de Groot MC, van Grootheest AC:
Proton pump inhibitor-induced acute interstitial nephritis. Br J Clin
trointestinal symptoms and often are treated with acid Pharmacol 64: 819 – 823, 2007
suppressive therapy with histamine2 (H2) receptor antag- 6. Sierra F, Suarez M, Rey M, Vela MF: Systematic review: Proton pump
inhibitor-associated acute interstitial nephritis. Aliment Pharmacol
onists and proton-pump inhibitors. H2 receptor antago- Ther 26: 545–553, 2007
nists are predominantly cleared by renal mechanisms that 7. Sica DA, Carl D, Zfass AM: Acute phosphate nephropathy: An
involve varying degrees of filtration and tubular secretion emerging issue. Am J Gastroenterol 102: 1844 –1847, 2007
8. Heher EC, Thier SO, Rennke H, Humphreys BD: Adverse renal and
by cationic transporters (3). On the basis of its ability to metabolic effects associated with oral sodium phosphate bowel prep-
block tubular secretion of creatinine, the first-generation aration. Clin J Am Soc Nephrol 3: 1494 –1503, 2008
H2 receptor antagonist cimetidine has been used as a way 9. Brunelli SM: Association between oral sodium phosphate bowel prep-
arations and kidney injury: a systematic review and meta-analysis.
to have 24-hour creatinine clearance measurements more Am J Kidney Dis 53: 448 – 456, 2009
closely approximate true GFR (4). The dosage of an H2
receptor antagonist is generally adjusted on the basis Antidiabetic Agents
of the level of renal function; however, concentration- Multiples therapies with differing mechanisms of
dependent adverse effects are uncommon with H2 action are available for the treatment of diabetes;
receptor antagonists other than cimetidine, which can however, in the patient with chronic kidney disease
cause bradyarrhythmias and confusion. (CKD) there are therapy considerations related to
Proton-pump inhibitors are predominantly hepati- dosage adjustment according to level of renal function,
cally cleared, and dosage adjustment is not necessary in and certain therapies are best avoided altogether (1).
the patient with CKD. As is the case with H2 receptor Exogenous insulin, whether it is short or long acting,
antagonists, sporadic cases of acute interstitial nephritis is mainly renally cleared by way of filtration and
have occurred with proton-pump inhibitors (5,6). Proton- extensive tubular secretion and cellular catabolism in
pump inhibitors as well as H2 receptor antagonists are the kidney. As renal failure progresses to GFR levels
available for over-the-counter purchase. As such, unless of less than approximately 30 ml/min, insulin clear-
a patient is specifically questioned about over-the- ance begins to decrease, its half-life increases, and
counter medication use, a potential cause of acute kidney insulin requirements begin to decline (2). As such,
disease can go unnoticed. hypoglycemic spells become more common unless
Acute phosphate nephropathy is a widely recog- there is some reduction in administered insulin dosage.
nized complication of the use of phosphate laxative Metformin is generally considered to be contra-
preparations, such as in patients who are preparing for indicated in advanced CKD because of the associated
colonoscopy. Advanced age, renal failure, and use of risk for lactic acidosis. It can be used at low- to
medications such as angiotensin-converting enzyme mid-range dosages in patients with creatinine clear-
inhibitors and angiotensin-receptor blockers are rec- ances of 30 to 60 ml/min but should be avoided with
ognized as risk factors for the development of acute creatinine clearances of ⬍30 ml/min unless other ther-
phosphate nephropathy. The presence of any of these apy options are not available; however, few cases of
risk factors necessitates careful attention to avoiding metformin-associated lactic acidosis have been re-
excessive dehydration in the process of bowel cleans- ported, and most have occurred in patients with other
ing. In so doing, the likelihood that acute phosphate reasons for developing lactic acidosis, such as sepsis
nephropathy will occur can be reduced (7–9). or renal failure. There is considerable debate, there-
fore, as to the optimal use of metformin in patients
with advanced renal disease. This is further compli-
References cated by reliance on criteria for its use that are based
1. Maton PN, Burton ME: Antacids revisited: A review of their clinical
pharmacology and recommended therapeutic use. Drugs 57: 855– 870, on serum creatinine values (contraindicated for serum
1999 creatinine values ⬎1.5 mg/dl in men and ⬎1.4 mg/dl
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 263

in women) rather than actual or estimated GFR (3,4). There is no long-term safety information on the incre-
Although use of metformin continues to increase, tin-based secretagogues, such as sitagliptin. Sitagliptin
observational studies have not been able to demon- is renally cleared and requires dosage adjustment on the
strate an increased incidence of lactic acidosis in basis of the level of renal function; however, it is unclear
metformin-treated patients, even when it is used in what the risk of sitagliptin accumulation is (16).
populations with relative contraindications (5,6). Be- Treatment of the patient with late-stage renal
cause the use of metformin in the patient with CKD is failure and diabetes can prove difficult in that there are
in a state of conceptual evolution, the best approach is limitations and contraindications to the use of several
probably to assume that in patients without other of the therapies that routinely are used for this disease.
comorbid conditions that would predispose them to Even with the change in systemic clearance of insulin
lactic acidosis, elevated serum creatinine levels should in advanced CKD and ESRD, it often is the best option
be considered a risk factor for the development of in these patients to maintain glycemic control.
lactic acidosis but not an absolute contraindication to
the use of metformin. References
Although the metabolism of thiazolidinediones is 1. Snyder RW, Berns JS: Use of insulin and oral hypoglycemic medi-
cations in patients with diabetes mellitus and advance kidney disease.
unaffected by renal failure, they must be used with
Semin Dial 17: 365–370, 2004
caution in this context because of their volume-retain- 2. Biesenbach G, Raml A, Schmekal B, Eichbauer-Sturm G: Decreased
ing effects (7,8); however, in that the edema seen with insulin requirement in relation to GFR in nephropathic type 1 and
insulin-treated type 2 diabetic patients. Diabet Med 20: 642– 645, 2003
the thiazolidinediones is secondary to salt and water 3. Haneda M, Morikawa A: Which hypoglycaemic agents to use in type
retention, the volume-retaining effect of these com- 2 diabetic subjects with CKD and how? Nephrol Dial Transplant 24:
pounds is not a deterrent to their use in the patient with 338 –341, 2009
4. Mani MK: Metformin in renal failure: Weigh the evidence. Nephrol
ESRD. Dial Transplant 24: 2287–2288, 2009
First-generation sulfonylureas (acetohexamide, 5. Bodmer M, Meier C, Krähenbühl S, Jick SS, Meier CR: Metformin,
chlorpropamide, tolazamide, and tolbutamide) are sel- sulfonylureas, or other antidiabetes drugs and the risk of lactic
acidosis or hypoglycemia: A nested case-control analysis. Diabetes
dom used and should be avoided in patients with Care 31: 2086 –2091, 2008
stages 3 through 5 CKD. These agents were associated 6. Salpeter SR, Greyber E, Pasternak GA, Salpeter Posthumous EE: Risk of
with prolonged hypoglycemia relating to parent drug fatal and nonfatal lactic acidosis with metformin use in type 2 diabetes
mellitus. Cochrane Database Syst Rev (1): CD002967, 2010
and/or active metabolite accumulation (9,10). Among 7. Chapelsky MC, Thompson-Culkin K, Miller AK, Sack M, Blum R,
second-generation sulfonylureas, glibenclamide (gly- Freed MI: Pharmacokinetics of rosiglitazone in patients with varying
buride) is oxidized to several metabolites, one of degrees of renal insufficiency. J Clin Pharmacol 43: 252–259, 2003
8. Nesto RW, Bell D, Bonow RO, Fonseca V, Grundy SM, Horton ES,
which, 4-hydroxy-glibenclamide, is renally cleared Le Winter M, Porte D, Semenkovich CF, Smith S, Young LH, Kahn
and has 15% of the potency of the parent compound. R: Thiazolidinedione use, fluid retention, and congestive heart fail-
Thus, the risk for hypoglycemia is increased with its ure: A consensus statement from the American Heart Association and
American Diabetes Association. Diabetes Care 27: 256 –263, 2004
use in patients with renal dysfunction. Glipizide and 9. Harrower AD: Pharmacokinetics of oral antihyperglycaemic agents
gliclazide are metabolized by the liver to several inactive in patients with renal insufficiency. Clin Pharmacokinet 31: 111–
119, 1996
metabolites and thus generally are considered safe to be
10. Krepinsky J, Ingram AJ, Clase CM: Prolonged sulfonylurea-induced
used in patients with reduced renal function (11). hypoglycemia in diabetic patients with end-stage renal disease. Am J
The nonsulfonylurea insulin secretagogues repa- Kidney Dis 35: 500 –505, 2000
11. KDOQI: Clinical practice guidelines and clinical practice recommen-
glinide and nateglinide can be used in renal failure dations for diabetes and chronic kidney disease. Guideline 2: Man-
without dosage adjustments (11–13). A small amount agement of hyperglycemia and general diabetes care in chronic
of nateglinide is excreted in the urine, and its active kidney disease. Am J Kidney Dis 49[Suppl 2]: S62–S73, 2007
12. Schumacher S, Abbasi I, Weise D, Hatorp V, Sattler K, Sieber J,
metabolite is also renally excreted; there is a small risk Hasslacher C: Single- and multiple-dose pharmacokinetics of repa-
for hypoglycemia when this compound is used in glinide in patients with type 2 diabetes and renal impairment. Eur
advanced renal failure (14,15). ␣-Glucosidase inhibi- J Clin Pharmacol 57: 147–152, 2001
13. Lubowsky ND, Siegel R, Pittas AG: Management of glycemia in
tors such as acarbose and miglitol are generally not patients with diabetes mellitus and CKD. Am J Kidney Dis 50:
used in patients with advanced renal failure because 865– 879, 2007
the plasma level of the parent drug and variably active 14. Inoue T, Shibahara N, Miyagawa K, Itahana R, Izumi M, Nakanishi
T, Takamitsu Y: Pharmacokinetics of nateglinide and its metabolites
metabolites can accumulate, although the clinical sig- in subjects with type 2 diabetes mellitus and renal failure. Clin
nificant of such drug accumulation is not known (11). Nephrol 60: 90 –95, 2003
264 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

15. Nagai T, Imamura M, Iizuka K, Mori M: Hypoglycemia due to (8,9). Codeine, although commonly thought of as a
nateglinide administration in diabetic patient with chronic renal
failure. Diabetes Res Clin Pract 59: 191–194, 2003
“mild” narcotic analgesic, also presents problems in the
16. Bergman AJ, Cote J, Yi B, Marbury T, Swan SK, Smith W, Gottes- patient with CKD. Codeine has multiple metabolites, the
diener K, Wagner J, Herman GA: Effect of renal insufficiency on the predominant one being codeine-6-glucoronide; both co-
pharmacokinetics of sitagliptin, a dipeptidyl peptidase-4 inhibitor.
Diabetes Care 30: 1862–1864, 2007
deine and codeine-6-glucoronide are renally excreted
(10). There is a report of respiratory arrest attributed to
Analgesic Agents the morphine-6-glucoronide metabolite of codeine (11).
Thus, morphine and codeine are best avoided in patients
Acetaminophen with significantly reduced GFR rather than trying to
There remains some debate on the role of long-term estimate the degree to which their dosage should be
acetaminophen use and the development of chronic kid- reduced on the basis of the level of renal function.
ney disease (CKD). Acetaminophen is the primary me- Normeperidine, a major metabolite of meperi-
tabolite of phenacetin. Despite the clearly established dine, has approximately 50% of the potency of me-
association between phenacetin and analgesic nephropa- peridine as an analgesic but two to three times its
thy, there is insufficient evidence to link habitual acet- potency as a neuroexcitatory agent and is predomi-
aminophen use to analgesic nephropathy (1,2); however, nantly renally cleared. Meperidine and normeperidine
the consistency of the results from existing epidemio- toxicities are not reversed by naloxone, and it may in
logic studies and the observed apparent dosage-response certain instances exacerbate it. Because of this, mepe-
relationship suggest that such a relationship cannot be dine use should be avoided in patients with significant
completely excluded. Among 1700 healthy women who renal dysfunction. Normeperidine is dialyzable; there-
participated in the Nurses’ Health Study in the United fore, hemodialysis may be used for patients with
States, those who consume ⬎3000 g of acetaminophen suspected normeperidine neurotoxicity (12).
had an odds ratio of 2.04 (95% confidence interval 1.28
to 3.24) for a decreased GFR of at least 30 ml/min per References
1.73 m2 during an 11-yr period compared with women 1. Barrett BJ: Acetaminophen and adverse chronic renal outcomes: An
appraisal of the epidemiologic evidence. Am J Kidney Dis 28[Suppl
who consumed ⱖ100 g (3). Nonetheless, single-sub- 1]: S14 –S19, 1996
stance acetaminophen and aspirin seem to be safe to use 2. Fored CM, Ejerblad E, Lindblad P, Fryzek JP, Dickman PW, Signo-
for patients with diagnosed advanced stages 4 to 5 CKD rello LB, Lipworth L, Elinder CG, Blot WJ, McLaughlin JK,
Zack MM, Nyrén O: Acetaminophen, aspirin, and chronic renal
without an adverse effect on CKD progression (4). failure. N Engl J Med 345: 1801–1808, 2001
3. Curhan GC, Knight EL, Rosner B, Hankinson SE, Stampfer MJ:
Opioid Analgesic Agents Lifetime nonnarcotic analgesic use and decline in renal function in
A growing body of literature now shows that mod- women. Arch Intern Med 164: 1519 –1524, 2004
erate to severe chronic pain is common in patients with 4. Evans M, Fored CM, Bellocco R, Fitzmaurice G, Fryzek JP,
McLaughlin JK, Nyrén O, Elinder CG: Acetaminophen, aspirin and
ESRD and affects virtually every aspect of health-related progression of advanced chronic kidney disease. Nephrol Dial Trans-
quality of life (5). A number of narcotic analgesic agents plant 24: 1908 –1918, 2009
5. Davison SN: Pain in hemodialysis patients: Prevalence, cause, sever-
can potentially be used in the management of pain in
ity, and management. Am J Kidney Dis 42: 1239 –1247, 2003
these patients, including morphine, codeine, hydromor- 6. Davies G, Kingswood C, Street M: Pharmacokinetics of opioids in
phone, and oxycodone, as well as fentanyl and metha- renal dysfunction. Clin Pharmacokinet 31: 410 – 422, 1996
7. Dean M: Opioids in renal failure and dialysis patients. J Pain
done. Fentanyl and methadone may be the safest agents Symptom Manage 28: 497–504, 2004
to use because they are hepatically metabolized and do 8. Conway BR, Fogarty DG, Nelson WE, Doherty CC: Opiate toxicity
not have active metabolites (6,7). in patients with renal failure. BMJ 332: 345–346, 2006
9. Sawe J, Odar-Cederlof I: Kinetics of morphine in patient with renal
Morphine use is problematic in the patient with failure. Eur J Clin Pharmacol 32: 377–382, 1987
CKD because it is hepatically metabolized to morphine- 10. Talbott GA, Lynn AM, Levy FH, Zelikovic I: Respiratory arrest
3-glucoronide (55%), morphine-6-glucuronide (10%), precipitated by codeine in a child with chronic renal failure. Clin
Pediatr 36: 171–173, 1997
and normorphine (4%), all of which are excreted renally, 11. Guay DR, Awni WM, Finlay JW, Halstenson CE, Abraham PA,
together with 10% of the parent compound. These active Opsahl JA, Jones EC, Matzke GR: Pharmacokinetics and pharmaco-
metabolites accumulate with repetitive dosing in the dynamics of codeine in end-stage renal disease. Clin Pharmacol Ther
43: 63–71, 1988
patient with CKD, with the attendant risk for respiratory 12. Hassan H, Bastani B, Gellens M: Successful treatment of normeperidine
depression and adverse central nervous system effects neurotoxicity by hemodialysis. Am J Kidney Dis 35: 146 –149, 2000
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

Nephrology Self-Assessment Program

Examination Questions
Instructions to obtain 8 AMA PRA Category 1 CreditsTM
Date of Original Release: July 2010
Examination Available Online: on or before Friday, July 9, 2010
Audio Files Available: No audio files will be available for this issue.
CME Credit Eligible Through: June 30, 2011
Answers: Correct answers with explanations will be posted on the ASN website in July 2011 when the issue is archived.
UpToDate Links Active: July and August 2010
Core Nephrology question links active: There are no core questions for this issue.
Target Audience: Nephrology Board and recertification candidates, practicing nephrologists, and internists.
Method of Participation:
● Read the syllabus that is supplemented by original articles in the reference lists, and complete the online self-assessment
examination.
● Examinations are available online only after the first week of the publication month. There is no fee. Each participant is
allowed two attempts to pass the examination (⬎75% correct).
● Your score and a list of question/s (by number) answered incorrectly can be printed immediately.
● Your CME certificate can be printed immediately after passing the examination.
● Answers and explanations are provided ONLY with a passing score on the first or second attempt.
● Your ASN transcript will be updated in 6 to 8 weeks after passing the examination.
Instructions to access the Online CME Center to take the examination and complete the evaluation:
● Login at ASN’s website: www.asn-online.org
● Click the “CME” tab at the top of the homepage
● In the left-hand column click “Online CME Center”
● Scroll down; in the center of the page click the computer screen image to “Visit the Online CME Center”
● Once you’re in the Center, click the “NephSAP” banner in the center of the page
● Select a topic and click on “Enter Activity” to the right
● Read through the instructions, and then click on “Continue” at the bottom of the page
● Click on “Examination Questions/Evaluations” to begin
● Your score and a list of question numbers answered incorrectly can be printed immediately
● Follow the prompts to retake the examination if you failed, or print your certificate and the correct answers if you passed
● You can save and finish the exam to complete at another time
● You can retake the exam at any time; each participant is allowed two attempts
Instructions to Obtain American Board of Internal Medicine (ABIM) Maintenance of Certification
(MOC) Points:
Each issue of NephSAP provides 10 MOC points. Respondents must meet the following criteria:
● Be certified by ABIM in internal medicine and/or nephrology and must be enrolled in the ABIM–MOC program
via the ABIM website (www.abim.org).
● Pass the self-assessment examination within the timeframe specified in this issue of NephSAP.
● Designate the issue for MOC points by clicking on the MOC link on the CME certificate page after passing the examination.
You will be leaving the ASN-CECity site and transferring the information directly to the ABIM in real-time.
● Provide your ABIM Certificate ID number and your date of birth.
● You will receive a confirmation message from the ABIM indicating the receipt of your information.
MOC points will be applied to only those ABIM candidates who have enrolled in the program. It is your responsibility to complete
the ABIM MOC enrollment process.

266
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

Volume 9, Number 4, July 2010 —Clinical Pharmacology for the Nephrologist

1. A 74-year-old man with blurred vision receives a simplify dosing, avoid toxicity, and thereby
diagnosis of fungal endophthalmitis, and Aspergillus minimize cost.
fumigatus is cultured from the vitreous aspirate. His
history is complicated with diabetes, severe chronic 3. The volume of distribution (Vd) of a drug can
be used to calculate both a loading dose and a
kidney disease (CKD; estimated GFR [eGFR] ⬍30),
maintenance dose and may demonstrate which
diabetic retinopathy, illicit intravenous drug use, hy-
ONE of the following?
pertension, and narcotic dependence. Intravenous vori-
conazole therapy is started; after 2 days of treatment, A. Vd increases with ascites, or “third
he complains to the home health agency that he is spacing.”
seeing bright “psychedelic” colors and “flashing B. Vd is not affected by uremic waste prod-
lights” even when his eyes are closed. His neurologic ucts and thus is irrelevant in ESRD.
and eye examinations are unremarkable. C. Expect greater drug removal by hemodial-
Which ONE of the following is the MOST ysis when Vd is large.
likely cause of these symptoms? D. Vd increases with volume contraction.
A. Undisclosed LSD use E. Changes in protein binding of a drug have
B. Diabetic retinopathy no impact on Vd.
C. Accumulation of voriconazole 4. A patient is begun on aminoglycoside therapy for
D. The solubilizing agent cyclodextrin, found the treatment of Gram-negative sepsis.
in voriconazole preparations Aminoglycoside nephrotoxicity correlates
E. Migraines with which ONE of the following?
A. Supratherapeutic doses given once per day
2. Which ONE of the following statements re- B. High peak and low trough levels
garding pharmacologic agents that undergo C. Frequency of dosing
hepatic metabolism and are not eliminated by
D. Post-antibiotic effect
the kidney is CORRECT?
E. Ototoxicity
A. Such drugs do not require dosage adjust-
ment in patients with CKD. 5. A 45-year-old man with ESRD of unknown cause and
B. Renal function may be relevant because of intermittent alcohol abuse receives hemodialysis thrice
renal elimination of active metabolites. weekly. He has had fistula stenosis detected recently,
and it required an increase in heparin use during his
C. Such agents do not induce the cytochrome
dialysis treatments. He is emergently admitted for
P450 enzyme system in patients with CKD to
upper gastrointestinal tract bleeding after his last dial-
the same degree seen in patients with intact
ysis treatment, but endoscopic findings are inconclu-
kidney function. sive. His medications include antihypertensive agents,
D. Such drugs do not inhibit the cytochrome P450 omeprazole, sevelamer carbonate, and sertraline for
enzyme system in patients with CKD to the depression. He denies recent increased use of his non-
same degree seen in patients with intact kidney steroidal anti-inflammatory drug as well as aspirin.
function. Which ONE of the following drugs is MOST
E. Such agents should always be chosen over likely to contribute to his episode of gastroin-
compounds that undergo renal elimination to testinal bleeding?
267
268 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

A. Increased heparin requirements at dialysis treat- C. Nifedipine is excreted through the kidney
ments and inhibits P450 3A4 gene family and
B. Sevelamer interacts with CsA.
C. None of his drugs would contribute to bleeding D. This is an iatrogenic drug toxicity most
likely as a result of drug interactions.
D. Sertraline
夝8. A 52-year-old man with diabetic nephropathy under-
6. A 32-year-old, 55-kg woman with end-stage renal
went a successful kidney transplant 7 months ago.
failure has undergone renal transplantation for
Except for one episode of rejection, which was treated
hypertensive nephropathy. Her urine output in the
with three pulse doses of intravenous methylpred-
last 24 hours ranges from 5 to 10 ml/hr. You
nisolone (500 mg), his posttransplantation course had
would like to start this patient on a cyclosporine
been unremarkable.
(CsA) immunosuppressive regimen but are con-
cerned about nephrotoxicity. His current medications include tacrolimus 2 mg
twice daily, mycophenolate mofetil 1000 mg twice
Which ONE of the following is the appropri-
daily, prednisone 15 mg/d, TMP/SMZ 1 DS tablet
ate treatment protocol for this patient?
three times weekly, lisinopril 10 mg/d, atorvastatin
A. Initiate CsA; renal function will return to 20 mg/d, NPH insulin 32 U in the morning and 17
normal in a few days. U in the evening, aspirin 81 mg/d, and a multivita-
B. Give 100 mg of furosemide, then start min.
CsA at 4 to 5 mg/kg per d in two divided He now presents with a 2-day history of a nonpro-
doses and monitor for toxicities. ductive cough, intermittent right-sided pleuritic chest
C. Consider the use of sirolimus at 10 mg/d. pain, temperature (today 103.5°F), and rigors. He
D. Consider an induction therapy with polyclonal has not traveled and has not had contacts with indi-
antibodies and delay the use of CsA for 3 to 5 viduals with respiratory illnesses. He has no history
days. of gardening or recent contacts with construction.
His social history was noncontributory.
7. A 32-year-old, 55-kg woman with end-stage renal Which ONE of the following drugs would be
failure underwent renal transplantation. Two months the BEST initial agent for the treatment of
later, she complains of a tingling sensation in her hand his pneumonia?
and severe hand tremors. Her weight has increased to
A. Azithromycin
65 kg, BP is 160/94 (baseline before discharge was
145/85), blood urea nitrogen level is 30, serum creat- B. Erythromycin
inine level is 2.2 mg/dl (baseline 1.2 mg/dl), and C. Clarithromycin
trough whole-blood (RIA monoclonal Incstar) CsA D. Antituberculous therapy
level is 885 ng/dl. Her medications include CsA 200
mg twice daily; lisinopril 10 mg/d; prednisone 20 9. Which ONE of the following pharmacologic
mg/d; fluconazole 400 mg/d; nifedipine 30 mg parameters is the MOST important one to
twice daily; and trimethoprim/sulfamethoxazole measure to adjust mycophenolate therapy?
(TMP/SMZ) double strength (DS) Monday, A. Mycophenolic acid (MPA) levels at 0, 2,
Wednesday, and Friday. and 6 hours
Which ONE of the following statements B. MPA/mycophenolate glucuronide concen-
BEST explains her symptoms? tration at trough
A. This presentation is consistent with acute C. Percentage of CD52 cells
rejection, and a biopsy is needed.
D. Pharmacokinetic monitoring of MPA or
B. Use of an angiotensin-converting enzyme mycophenolate glucuronide is not neces-
inhibitor (ACEI) for the treatment of hy- sary for most patients.
pertension could have contributed to her
renal dysfunction. 10. A 32-year-old woman received a renal transplant.
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 269

Her postoperative course was uneventful. Her serum medications include a diuretic, an ACEI, baby-
creatinine was 1.1 mg/dl at the time of discharge. Her dosage aspirin, and a statin.
medications at this time included tacrolimus 2 mg Which ONE of the following is indicated at
twice daily; sirolimus 3 mg/d; TMP/SMZ 1 DS tablet this time?
Monday, Wednesday, and Friday; lisinopril 10 mg/d;
A. Continue with the recommended therapy.
simvastatin 40 mg/d; and aspirin 81 mg/d. During the
past 12 months, her renal function has progressively B. Continue with the recommended bowel prep-
worsened. Her serum creatinine is 2.0 mg/dl today, aration, but discontinue her diuretic and
and a biopsy did not reveal rejection but is consistent ACEI at least 2 days before the procedure.
with signs of tacrolimus nephrotoxicity. C. Hospitalize her for the bowel preparation,
Which ONE of the following immunosuppres- and provide intravenous fluid replacement.
sive agents has been used alone to decrease D. Recommend the use of polyethylene glycol
nephrotoxicity? as the bowel-preparation agent.
A. Sirolimus
14. A 68-year-old man with diabetes and stage 3 CKD
B. Cyclosporine has a serum creatinine level of 2.0 mg/dl, an eGFR of
C. Tacrolimus 40 ml/min, and significant peripheral edema. He is
D. Cyclophosphamide being seen for a routine office visit, and he states that
he has recently seen a deterioration in his diabetes
夝11. Which ONE of the following is the dose- control with a most recent hemoglobin A1c of 8.0%
limiting toxicity of sirolimus? compared with a previous value of 6.5%. The only
A. Hyperlipidemia medication change had been the discontinuation of
his metformin by his primary care physician because
B. Acute rejection
his serum creatinine level was elevated and that it
C. Hypertension might be “damaging the kidney.” In its place, his
D. Gastrointestinal complications sulfonylurea dosage was doubled because he was
hesitant to start insulin.
12. A 23-year-old female kidney transplant recipient de- Which ONE of the following is the BEST
velops severe hirsutism after kidney transplantation. advice that can be given to him about his
Which ONE of the following agents could diabetes management?
replace cyclosporine for maintenance ther- A. Increase his dosage of the sulfonylurea,
apy to avoid cosmetic adverse effects such and recheck glycemic status in 2 weeks.
as hirsutism and coarsening of facial fea-
B. Add a thiazolidinedione to the sulfonylurea.
tures?
C. Restart the metformin at a reduced dosage, and
A. Thymoglobulin
recheck his glycemic status in several weeks.
B. Tacrolimus
D. Discontinue oral medications and instead begin
C. Sirolimus a regimen of short- and long-acting insulin.
D. Mycophenolate
夝15. Which ONE of the following statements is
夝13. A 78-year-old woman is scheduled for a screening CORRECT regarding the use of a thiazo-
colonoscopy, and her gastroenterologist has recom- lidinedione in a patient with stage 4 CKD?
mended a standard bowel preparation protocol in- A. Increased risk for lactic acidosis
cluding a laxative and neutral sodium phosphate
B. Increased risk for developing significant
orally. She received a similar bowel-cleansing
peripheral edema
regimen 2 years ago without incident. At that
time, an adenomatous colonic polyp was re- C. Increased risk for hypoglycemic episodes
moved. Her serum creatinine at its baseline is D. Acceleration in the rate of renal functional
1.5 mg/dl, and her eGFR is 52 ml/min. Her decline
270 Nephrology Self-Assessment Program - Vol 9, No 4, July 2010

16. A 38-year-old man with an eGFR of 24 ml/min and dialysis, and has just been started on ranolazine
diabetic nephropathy is being seen for recurrent hy- for management of angina.
poglycemic spells. His medication regimen includes Which ONE of the following statements is
a first-generation sulfonylurea, a loop diuretic, and an CORRECT regarding ranolazine therapy in
ACEI. this patient?
Which ONE of the following should be done A. Dosing is not an issue in that it is hepati-
to prevent additional hypoglycemic spells? cally cleared.
A. Change him from an ACEI to an angioten- B. Ranolazine has a narrow therapeutic window,
sin receptor blocker. and its clearance is diminished in renal disease.
B. Increase carbohydrate intake. C. Ranolazine has a narrow therapeutic win-
C. Convert from a first-generation to a sec- dow and can cause acute kidney injury.
ond-generation sulfonylurea. D. Ranolazine produces a reversible 10 to
D. Reduce his dosage of sulfonylurea. 15% reduction in eGFR.
E. Discontinue the sulfonylurea and begin an
insulin regimen. 20. A 64-year-old woman with ESRD that is treated with
hemodialysis develops significant peripheral edema
upon recently starting low-dosage amlodipine. She
17. Which ONE of the following BEST relates to has residual renal function of 4 ml/min as determined
the use of acetaminophen in stage 4 to 5 by creatinine clearance and states that the amlodipine
CKD? has helped lower her interdialytic home BP readings
A. Acetaminophen use accelerates decline in from 150/86 to 138/78 mmHg.
GFR in 20% of patients. Which ONE of the following recommenda-
B. Acetaminophen use leads to an increased tions is CORRECT?
risk for diuretic resistant hypertension. A. Gradually reduce the patient’s target weight on
C. Acetaminophen is the preferred first-line dialysis until the edema has resolved.
nonnarcotic analgesic. B. Convert to another channel blocker, such
D. Acetaminophen dosing requires adjustment as verapamil or diltiazem.
for level of eGFR. C. Start high-dosage loop diuretic therapy.
D. Stop the amlodipine and restart after sev-
18. A 58-year-old man with ESRD secondary to eral days at a lower dosage.
polycystic kidney disease is seen by his primary
care physician and started on simvastatin be- 夝21. A 34-year-old patient with ESRD from FSGS is
cause of his presumed high cardiovascular risk. found at most of his dialysis sessions to enter with
Approximately 3 weeks thereafter, he develops marked hypertension. His last treatment was charac-
atrial fibrillation and is placed on diltiazem for terized by a BP of 190/110 mmHg despite his achiev-
rate control. He subsequently develops myalgias ing near target weight at entry to the session. Discus-
and muscle weakness. sion with his wife suggests that he has not been
Which ONE of the following is the BEST adherent to his antihypertensive medication regimen.
treatment approach for this patient? Which ONE of the following would you
A. Continue current therapy, but add coen- now recommend?
zyme Q. A. Transdermal clonidine
B. Convert from diltiazem to verapamil. B. Performing pill counts at each dialysis
C. Switch from simvastatin to lovastatin. treatment visit
D. Discontinue the simvastatin. C. Consulting a home health care agency to
arrange medication education at home
19. A male patient has stage 5 CKD, is not yet on D. Home BP monitoring
Nephrology Self-Assessment Program - Vol 9, No 4, July 2010 271

22. A 27-year-old patient with stage 4 CKD from FSGS C. Increased plasma protein binding of torsemide
is found to have a BP of 190/110 mm Hg. You elect D. Reduced delivery of torsemide to the as-
to add atenolol 50 mg/d to his treatment program of cending limb
an ACEI and a dihydropyridine calcium channel
E. His degree of cardiac compromise
blocker, each at maximal dosage. Within 1 week, he
develops two episodes of bradycardia to 48 bpm. 24. A 45-year-old man with ischemic cardiomyopathy
When seen in the emergency department for light- presents with hyperkalemia of 6.2 mEq/L. He had
headedness, an electrocardiogram showed first-de- always manifested normal serum potassium levels.
gree heart block with sinus bradycardia. Serum elec- He recently had a medication change in which he
trolytes, calcium, and magnesium values all were in was started on a new combination of drugs. His
the normal range. serum creatinine level is 2.1 mg/dl and has been
Which ONE of the following statements stable for the past 12 months.
BEST explains the cause of his clinical dis- Which ONE of the following new drug com-
order? binations is MOST likely to have led to his
A. Idiosyncratic response to atenolol hyperkalemia?
B. Reduced atenolol clearance A. Eplerenone and diltiazem
C. Atenolol interaction with a dihydropyridine B. Eplerenone and metalozone
calcium channel C. Spironolactone and torsemide
D. A genetic polymorphism in his hepatic D. Spironolactone and acetazolamide
metabolism (CYP3A4) of atenolol
25. A 47-year-old man with stage 4 CKD secondary to
diabetic nephropathy has had progressive episodes of
23. A 44-year-old patient with stage 4 CKD (eGFR 22
vomiting. A gastrointestinal evaluation determined
ml/min) as a result of hypertension and New York
that the patient had gastroparesis and was begun on
Heart Association class 2 congestive heart failure has
metoclopramide. During the subsequent 2 weeks, he
developed increased peripheral edema. He had been
complained of muscle pain and manifested rigidity,
treated with a loop diuretic, in addition to his cardiac
reduced reflexes, hyperthermia, and confusion. He
regimen of an ACEI and eplerenone. He had been
was admitted to the hospital and found to have the
receiving increasing dosages of furosemide but has
following abnormalities on physical examination:
noted less response after an 80-mg dose. You decide
Temperature of 101°F and lethargy.
to switch his diuretic to torsemide, yet he remains
diuretic resistant. Which ONE of the following treatments
would you recommend at this time?
Which ONE of the following BEST explains
his diuretic resistance? A. Hemodialysis
A. Increased hepatic clearance of torsemide in B. Continuous hemofiltration
CKD C. Bromocriptine
B. Reduced filtration of torsemide D. Cyproheptadine

Você também pode gostar